Future Visioning The Role Of CRISPR Gene Editing: Navigating Law And Ethics To Regenerate Health And Cure Disease – marketscreener.com
"Despite the projected growth in market applications and abundant investment capital, there is a danger that legal and ethical concerns related to genetic research could put the brakes on gene editing technologies and product programs emanating therefrom."
What is Genome Editing?
There are thousands of diseases occurring in humans, animals, and plants caused by aberrant DNA sequences. Traditional small molecule and biologic therapies have only had minimal success in treating many of these diseases because they mitigate symptoms while failing to address the underlying genetic causes. While human understanding of genetic diseases has increased tremendously since the mapping of the human genome in the late 1990s, our ability to treat them effectively has been limited by our historical inability to alter genetic sequences.
The science of gene editing was born in the 1990s, as scientists developed tools such as zinc-finger nucleases (ZFNs) and TALE nucleases (TALENs) to study the genome and attempt to alter sequences that caused disease. While these systems were an essential first step to demonstrate the potential of gene editing, their development was challenging in practice due to the complexity of engineering protein-DNA interactions.
Then, in 2011, Dr. Emmanuelle Charpentier, a French professor of microbiology, genetics, and biochemistry, and Jennifer Doudna, an American professor of biochemistry, pioneered a revolutionary new gene-editing technology called CRISPR/Cas9. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and Cas9 stands for CRISPR-associated protein 9. In 2020, the revolutionary work of Drs. Charpentier and Doudna developing CRISPR/Cas9 were recognized with the Nobel Prize for Chemistry. The technology was also the source of a long-running and high-profile patent battle between two groups of scientsists.
CRISPR/Cas9 for gene editing came about from a naturally occurring viral defense mechanism in bacteria. The system is cheaper and easier to use than previous technologies. It delivers the Cas9 nuclease complexed with a synthetic guide RNA (gRNA) into a cell, cutting the 'cell's genome at the desired location, allowing existing genes to be removed and new ones added to a living organism's genome. The technique is essential in biotechnology and medicine as it provides for the genomes to be edited in vivo with extremely high precision, efficiently, and with comparative ease. It can create new drugs, agricultural products, and genetically modified organisms or control pathogens and pests. More possibilities include the treatment of inherited genetic diseases and diseases arising from somatic mutations such as cancer. However, its use in human germline genetic modification is highly controversial.
The following diagram from CRISPR Therapeutics AG, a Swiss company, illustrates how it functions:
Market Applications
In the 1990s, nanotechnology and gene editing were necessary plot points for science fiction films. In 2020, developments like nano-sensors and CRISPR gene editing technology have moved these technologies directly into the mainstream, opening a new frontier of novel market applications. According to The Business Research Company, the global CRISPR technology market reached a value of nearly $700 million in 2019, is expected to more than double in 2020, and reach $6.7 billion by 2030. Market applications target all forms of life, from animals to plants to humans.
Gene editing's primary market applications are for the treatment of genetically-defined diseases. CRISPR/Cas9 gene editing promises to enable the engineering of genomes of cell-based therapies and make them safer and available to a broader group of patients. Cell therapies have already begun to make a meaningful impact on specific diseases, and gene editing helps to accelerate that progress across diverse disease areas, including oncology and diabetes.
In the area of human therapy, millions of people worldwide suffer from genetic conditions. Gene-editing technologies like CRISPR-Cas9 have introduced a way to address the cause of debilitating illnesses like cystic fibrosis and create better interventions and therapies. They also have promising market applications for agriculture, food safety, supply, and distribution. For example, grocery retailers are even looking at how gene editing could impact the products they sell. Scientists have created gene-edited crops like non-browning mushrooms and mildew-resistant grapes - experiments that are part of an effort to prevent spoilage, which could ultimately change the way food is sold.
Investment Capital
Despite the inability to travel and conduct face-to-face meetings, attend industry conferences or conduct business other than remotely or with social distance, the investment markets for venture, growth, and private equity capital, as well as corporate R&D budgets, have remained buoyant through 2020 to date. Indeed, the third quarter of 2020 was the second strongest quarter ever for VC-backed companies, with 88 companies raising rounds worth $100 million or more according to the latest PwC/Moneytree report. Healthcare startups raised over $8 billion in the quarter in the United States alone. Gene-editing company Mammouth Biosciences raised a $45 million round of Series B capital in the second quarter of 2020. CRISPR Therapeutics AG raised more in the public markets in primary and secondary capital.
Bayer, Humboldt Fund and Leaps are co-leading a $65 million Series A round for Metagenomi, a biotech startup launched by UC Berkeley scientists. Metagenomi, which will be run by Berkeley's Brian Thomas, is developing a toolbox of CRISPR- and non-CRISPR-based gene-editing systems beyond the Cas9 protein. The goal is to apply machine learning to search through the genomes of these microorganisms, finding new nucleases that can be used in gene therapies. Other investors in the Series A include Sozo Ventures, Agent Capital, InCube Ventures and HOF Capital. Given the focus on new therapies and vaccines to treat the novel coronavirus, we expect continued wind in the sails for gene-editing companies, particularly those with strong product portfolios that leverage the technology.
Legal and Ethical Considerations
Despite the projected growth in market applications and abundant investment capital, there is a danger that legal and ethical concerns related to genetic research could put the brakes on gene-editing technologies and product programs emanating therefrom. The possibility of off-target effects, lack of informed consent for germline therapy, and other ethical concerns could cause government regulators to put a stop on important research and development required to cure disease and regenerate human health.
Gene-editing companies can only make money by developing products that involve editing the human genome. The clinical and commercial success of these product candidates depends on public acceptance of gene-editing therapies for the treatment of human diseases. Public attitudes could be influenced by claims that gene editing is unsafe, unethical, or immoral. Consequently, products created through gene editing may not gain the acceptance of the government, the public, or the medical community. Adverse public reaction to gene therapy, in general, could result in greater government regulation and stricter labeling requirements of gene-editing products. Stakeholders in government, third-party payors, the medical community, and private industry must work to create standards that are both safe and comply with prevailing ethical norms.
The most significant danger to growth in gene-editing technologies lies in ethical concerns about their application to human embryos or the human germline. In 2016, a group of scientists edited the genome of human embryos to modify the gene for hemoglobin beta, the gene in which a mutation occurs in patients with the inherited blood disorder beta thalassemia. Although conducted in non-viable embryos, it shocked the public that scientists could be experimenting with human eggs, sperm, and embryos to alter human life at creation. Then, in 2018, a biophysics researcher in China created the first human genetically edited babies, twin girls, causing public outcry (and triggering government sanctioning of the researcher). In response, the World Health Organization established a committee to advise on the creation of standards for gene editing oversight and governance standards on a global basis.
Some influential non-governmental agencies have called for a moratorium on gene editing, particularly as applied to altering the creation or editing of human life. Other have set forth guidelines on how to use gene-editing technologies in therapeutic applications. In the United States, the National Institute of Health has stated that it will not fund gene-editing studies in human embryos. A U.S. statute called "The Dickey-Wicker Amendment" prohibits the use of federal funds for research projects that would create or destroy human life. Laws in the United Kingdom prohibit genetically modified embryos from being implanted into women. Still, embryos can be altered in research labs under license from the Human Fertilisation and Embryology Authority.
Regulations must keep pace with the change that CRISPR-Cas9 has brought to research labs worldwide. Developing international guidelines could be a step towards establishing cohesive national frameworks. The U.S. National Academy of Sciences recommended seven principles for the governance of human genome editing, including promoting well-being, transparency, due care, responsible science, respect for persons, fairness, and transnational co-operation. In the United Kingdom, a non-governmental organization formed in 1991 called The Nuffield Council has proposed two principles for the ethical acceptability of genome editing in the context of reproduction. First, the intervention intends to secure the welfare of the individual born due to such technology. Second, social justice and solidarity principles are upheld, and the intervention should not result in an intensifying of social divides or marginalizing of disadvantaged groups in society. In 2016, in application of the same, the Crick Institute in London was approved to use CRISPR-Cas9 in human embryos to study early development. In response to a cacophony of conflicting national frameworks, the International Summit on Human Gene Editing was formed in 2015 by NGOs in the United States, the United Kingdom and China, and is working to harmonize regulations global from both the ethical and safety perspectives. As CRISPR co-inventor Jennifer Doudna has written in a now infamous editorial in SCIENCE, "stakeholders must engage in thoughtfully crafting regulations of the technology without stifling it."
Where Do We Go from Here?
The COVID-19 pandemic has forced us to rely more on new technologies to keep us healthy, adapt to working from home, and more. The pandemic makes us more reliant on innovative digital, biological, and physical solutions. It has created a united sense of urgency among the public and private industry (together with government and academia) to be more creative about using technology to regenerate health. With continued advances in computing power, network architecture, communications bandwidths, artificial intelligence, machine learning, and gene editing, society will undoubtedly find more cures for debilitating disease and succeed in regenerating human health. As science advances, it inevitably intersects with legal and ethical norms, both for individuals and civil society, and there are new externalities to consider. Legal and ethical norms will adapt, rebalancing the interests of each. The fourth industrial revolution is accelerating, and hopefully towards curing disease.
Originally published by IPWatchdog.com, November 24, 2020.
The content of this article is intended to provide a general guide to the subject matter. Specialist advice should be sought about your specific circumstances.
Mr Louis LehotL2 Counsel407 California Avenue, Suite #2Palo AltoCaliforniaUNITED STATESTel: 6507967280E-mail: louis.lehot@L2counsel.comURL: http://www.l2counsel.com/
Mondaq Ltd, 2020 - Tel. +44 (0)20 8544 8300 - http://www.mondaq.com, source Business Briefing
Read this article:
Future Visioning The Role Of CRISPR Gene Editing: Navigating Law And Ethics To Regenerate Health And Cure Disease - marketscreener.com
Recommendation and review posted by Bethany Smith
The Future of Global Gene Therapy Market To Observe Exponential Growth By 2020-2027 | Reports Globe – Cheshire Media
Fort Collins, Colorado The The Future of Global Gene Therapy Market 2020 Research Report provides information on the market size, share, trends, growth, competitive landscape, challenges and opportunities, revenue, and forecast to 2027. Reports Globe recently incorporated a comprehensive overview of the The Future of Global Gene Therapy market into its extensive database. The The Future of Global Gene Therapy Market report has been aggregated by collecting informative data on various dynamics such as market drivers, restraints, and opportunities.
This innovative report uses SWOT, PESTLE, and Porters Five Forces analysis to get a deeper look at the The Future of Global Gene Therapy market.
Download Exclusive Sample PDF along with few company profiles @ https://reportsglobe.com/download-sample/?rid=21308
The Major Players Profiled in this Report include
Impact of Covid-19 on this Market:
The coronavirus pandemic (COVID-19) has affected all aspects of life around the world. The study provides a comprehensive overview of the impact of the COVID-19 pandemic on the The Future of Global Gene Therapy market and its key segments. It also covers the current and future impact of the pandemic and offers a post-COVID-19 scenario to provide a deeper understanding of the dynamic changes in market trends and scenarios.
Competitive Landscape:
Competitive Analysis is one of the best sections of the report, comparing the progress of leading companies using key metrics like market share, new developments, global reach, local competition, pricing, and production. . From the nature of the competition to future changes in the vendor landscape, the report offers an in-depth competitive analysis in the global The Future of Global Gene Therapy Market.
Request a Discount on the report @ https://reportsglobe.com/ask-for-discount/?rid=21308
Market Segments and Sub-segments Covered in the Report are as per below:
Research Methodology:
Reports Globe follows a comprehensive research methodology to provide the most accurate market analysis. The company relies on a data triangulation model that can be used to assess market dynamics and provide accurate estimates. Key elements of the research methodology used for all of our market reports include:
Request customization of the report @https://reportsglobe.com/need-customization/?rid=21308
In addition, Reports Globe has access to a wide range of reputable regional and global paid databases that help the company understand regional and global market trends and dynamics. The company analyzes the industry from a 360-degree perspective, i.e. H. From the supply and demand side, which allows us to provide detailed information about the entire ecosystem for each study. Finally, a top-down and bottom-up approach is taken to get the final search results.
It includes analysis on the following
Finally, the The Future of Global Gene Therapy Market Report is a credible source of market research that will accelerate your business exponentially. The report gives the most important regional framework conditions, economic situations with item value, advantage, limit, production, supply, demand, market development rate and number, etc. The Future of Global Gene Therapy Industry Report Also includes a new SWOT review task, speculative test research, and corporate return on investment research.
To learn more about the report, visit @ https://reportsglobe.com/product/the-future-of-global-gene-therapy-market/
Thanks for reading this article; you can also get individual chapter wise section or region wise report versions like North America, Europe, or Asia.
How Reports Globe is different than other Market Research Providers
The inception of Reports Globe has been backed by providing clients with a holistic view of market conditions and future possibilities/opportunities to reap maximum profits out of their businesses and assist in decision making. Our team of in-house analysts and consultants works tirelessly to understand your needs and suggest the best possible solutions to fulfill your research requirements.
Our team at Reports Globe follows a rigorous process of data validation, which allows us to publish reports from publishers with minimum or no deviations. Reports Globe collects, segregates, and publishes more than 500 reports annually that cater to products and services across numerous domains.
Contact us:
Mr. Mark Willams
Account Manager
US: +1-970-672-0390
Email:[emailprotected]
Web:reportsglobe.com
See original here:
The Future of Global Gene Therapy Market To Observe Exponential Growth By 2020-2027 | Reports Globe - Cheshire Media
Recommendation and review posted by Bethany Smith
Global Prostate Cancer Therapeutics Market Size And Forecast (2020-2026)| With Post Impact Of Covid- – PharmiWeb.com
DBMR has added a new report titled Global Prostate Cancer Therapeutics Market with data Tables for historical and forecast years represented with Chats & Graphs spread through Pages with easy to understand detailed analysis. The Global Prostate Cancer Therapeutics Market report provides the list of leading competitors, strategic industry analysis and the insights of key factors influencing the industry. The market analysis and insights included in this Global Prostate Cancer Therapeutics Market research report offers key statistics on the market status of global and regional manufacturers and is an imperative source of guidance which provides right direction to the companies and individuals interested in the industry. This Global Prostate Cancer Therapeutics Market report is also all-embracing of the data which includes market definition, classifications, applications, engagements, market drivers and market restraints that are obtained with the help of SWOT analysis.
Market drivers and market restraints explained in this Global Prostate Cancer Therapeutics Market business research report provides idea about the rise or fall in the consumer demand for the particular product depending on several factors. Thoroughly described market segmentation aspect provides a clear idea about the product consumption based on several factors ranging from type, application, deployment model, end user to geographical region. This Global Prostate Cancer Therapeutics Market report also presents an analysis of prime manufacturers, trends, opportunities, marketing strategies, market effect factor and consumer needs by major regions, types, and applications globally while considering the past, present and future state of the industry.
Prostate cancer therapeutics market is expected to gain market growth in the forecast period of 2020 to 2027. Data Bridge Market Research analyses the market to reach a market value of USD 18.71 billion by 2027 while growing at a CAGR of 7.70% in the above-mentioned forecast period. Prostate cancer therapeutics market is growing due to factor such as increasing cases of prostate cancer and cardiovascular diseases.
Get Sample Report + All Related Graphs & Charts @https://www.databridgemarketresearch.com/request-a-sample?dbmr=global-prostate-cancer-therapeutics-market
Some of the major players operating in the global prostate cancer therapeutics market are Tolmar INC, Ferring Pharmaceuticals, Takeda pharmaceutical co. Ltd., Teva Pharmaceutical Industries LTD, Sanofi-Aventis, Pfizer Inc, Johnson & Johnson, IPSEN, Endo Pharmaceuticals Inc (Indevus Pharmaceuticals Inc) , Dendreon Corporation, Bristol-Myers Squibb, Astellas Pharma Inc, Astrazeneca Plc, Active Biotech, Abbott Laboratories, Bayer AG, Dendreon Corporation (Sanpower Group Co. Ltd.), AbbVie, Inc. among others.
Global Prostate Cancer Therapeutics Market By Drug Class (Hormonal Therapy, Chemotherapy, Immunotherapy, Targeted Therapy), Distribution Channel (Hospital Pharmacies, Retail Pharmacies, Online Sales, Others), Country (U.S., Canada, Mexico, Germany, Italy, U.K., France, Spain, Netherland, Belgium, Switzerland, Turkey, Russia, Hungary, Lithuania, Austria, Ireland, Norway, Poland, Rest of Europe, Japan, China, India, South Korea, Australia, Singapore, Malaysia, Thailand, Indonesia, Philippines, Vietnam, Rest of Asia- Pacific, Brazil, Peru, Argentina, Rest of South America, South Africa, Saudi Arabia, UAE, Kuwait, Egypt, Israel, Rest of Middle East & Africa), Market Trends and Forecast to 2027
Market Definition:Global Prostate Cancer Therapeutics Market
The prostate cancer occurs in the prostate which is a small walnut shaped gland. The prostate cancer is most common type in men. Due to increase in awareness regarding the symptoms among the people, the market for the prostate cancer therapeutics is growing at a high growth rate. Various developments in advance science are helping in development of launch of various options for the treatment of this disease. According to an article published recently by the cancer research institute in U.K., around 11,287 deaths were registered due to prostate cancer in U.K. The prostate cancer is the fourth most prevalent cancer globally. Various researches are made by the key player for the development of the therapies for the treatment of the cancer. The government is also taking various measures for the awareness regarding symptoms of the prostate cancer and availability of screening & diagnostic tests such as Prostate-Specific Antigen (PSA) and Digital Rectal Exam (DRE) resulting in early detection. Hence, such initiatives by the government and the key players help in the growth of the market.
Increasing awareness among people regarding prostate cancer along with technological advancement in screening and diagnostic tests, rising preferences of healthy living will increased geriatric population will enhance the growth of the prostate cancer therapeutics market in the forecast period of 2020-2027. On the other hand, limited number of players, increasing pharmaceutical expenditure will further create new opportunities for the growth of prostate cancer therapeutics market in the above mentioned forecast period.
Increasing cost of the treatment and adverse impact of treatment along with less success rate will acts as a restraint factor for the growth of prostate cancer therapeutics market in the above mentioned forecast period.
Competitive Landscape and Prostate Cancer Therapeutics Market Share Analysis
Prostate cancer therapeutics market competitive landscape provides details by competitor. Details included are company overview, company financials, revenue generated, market potential, investment in research and development, new market initiatives, global presence, production sites and facilities, production capacities, company strengths and weaknesses, product launch, product width and breadth, application dominance. The above data points provided are only related to the companies focus related to prostate cancer therapeutics market.
Market Segmentation:Global Prostate Cancer Therapeutics Market
The global prostate cancer therapeutics market is segmented based on drug type, distribution channel and geographical segments.
On the basis of drug type, the market is classified into hormonal therapy, luteinizing hormone-releasing hormone (LHRH) agonist, anti-androgens, immunotherapy, targeted therapy, and chemotherapy
On the basis of distribution channel, the market is segmented into hospital pharmacies, retail pharmacies, and online pharmacies.
Based on geography, the global prostate cancer therapeutics market report covers data points for 28 countries across multiple geographies namely North America & South America, Europe, Asia-Pacific and, Middle East & Africa. Some of the major countries covered in this report are U.S., Canada, Germany, France, U.K., Netherlands, Switzerland, Turkey, Russia, China, India, South Korea, Japan, Australia, Singapore, Saudi Arabia, South Africa and, Brazil among others.
Key Developments in the Market:
Grab Your Report at an Impressive 30% Discount! Please click Here @https://www.databridgemarketresearch.com/inquire-before-buying?dbmr=global-prostate-cancer-therapeutics-market
Prostate Cancer Therapeutics Market Country Level Analysis
Prostate cancer therapeutics market is analysed and market size insights and trends are provided by country, drug class and distribution channel as referenced above.
The countries covered in the prostate cancer therapeutics market report are U.S., Canada and Mexico in North America, Germany, France, U.K., Netherlands, Switzerland, Belgium, Russia, Italy, Spain, Turkey, Hungary, Lithuania, Austria, Ireland, Norway, Poland, Rest of Europe in Europe, China, Japan, India, South Korea, Singapore, Malaysia, Australia, Thailand, Indonesia, Philippines, Vietnam, Rest of Asia-Pacific (APAC) in the Asia-Pacific (APAC), Saudi Arabia, U.A.E, South Africa, Kuwait, Egypt, Israel, Rest of Middle East and Africa (MEA) as a part of Middle East and Africa (MEA), Peru, Brazil, Argentina and Rest of South America as part of South America.
North America dominates the prostate cancer therapeutics market due to increasing occurrence of prostate cancer and high mortality rate along with rising investment in research and development of advanced solutions while Asia-Pacific will witness a growth in forecast period of 2020-2027 because of increasing awareness among people regarding tumours and improvement in healthcare infrastructure in this regions.
The country section of the prostate cancer therapeutics market report also provides individual market impacting factors and changes in regulation in the market domestically that impacts the current and future trends of the market. Data points such as consumption volumes, production sites and volumes, import export analysis, price trend analysis, cost of raw materials, down-stream and upstream value chain analysis are some of the major pointers used to forecast the market scenario for individual countries. Also, presence and availability of global brands and their challenges faced due to large or scarce competition from local and domestic brands, impact of domestic tariffs and trade routes are considered while providing forecast analysis of the country data.
Patient Epidemiology Analysis
Prostate cancer therapeutics market also provides you with detailed market analysis for patient analysis, prognosis and cures. Prevalence, incidence, mortality, adherence rates are some of the data variables that are available in the report. Direct or indirect impact analysis of epidemiology to market growth are analysed to create a more robust and cohort multivariate statistical model for forecasting the market in the growth period.
Major Market Drivers and Restraints:
Reasons to Purchase this Report
Customization of the Report
About Data Bridge Market Research:
Data Bridge Market Research set forth itself as an unconventional and neoteric Market research and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best market opportunities and foster efficient information for your business to thrive in the market. Data Bridge endeavors to provide appropriate solutions to the complex business challenges and initiates an effortless decision-making process.
Contact:
Data Bridge Market Research
US: +1 888 387 2818
UK: +44 208 089 1725
Hong Kong: +852 8192 7475
Email: Corporatesales@databridgemarketresearch.com
This content has been distributed via CDN Newswire press release distribution service. For press release enquires please mail us at contact@cdnnewswire.com.
Recommendation and review posted by Bethany Smith
Cancer Gene Therapy Market Research Report with Revenue, Gross Margin, Market Share and Future Prospects till 2026 – Cheshire Media
The Cancer Gene Therapy Market grew in 2019, as compared to 2018, according to our report, Cancer Gene Therapy Market is likely to have subdued growth in 2020 due to weak demand on account of reduced industry spending post Covid-19 outbreak. Further, Cancer Gene Therapy Market will begin picking up momentum gradually from 2021 onwards and grow at a healthy CAGR between 2021-2025
Deep analysis about market status (2016-2019), competition pattern, advantages and disadvantages of products, industry development trends (2019-2025), regional industrial layout characteristics and macroeconomic policies, industrial policy has also been included. From raw materials to downstream buyers of this industry have been analysed scientifically. This report will help you to establish comprehensive overview of the Cancer Gene Therapy Market
Get a Sample Copy of the Report at: https://i2iresearch.com/report/global-cancer-gene-therapy-market-2020-market-size-share-growth-trends-forecast-2025/#download-sample
The Cancer Gene Therapy Market is analysed based on product types, major applications and key players
Key product type:Oncolytic VirotherapyGene TransferGene-Induced Immunotherapy
Key applications:HospitalsDiagnostics CentersResearch Institutes
Key players or companies covered are:AdaptimmuneBluebird bioCelgeneShanghai Sunway BiotechShenzhen SiBiono GeneTechSynerGene TherapeuticsAltor BioScienceAmgenArgenxBioCancellGlaxoSmithKlineMerckOncoGenex PharmaceuticalsTransgene
The report provides analysis & data at a regional level (North America, Europe, Asia Pacific, Middle East & Africa , Rest of the world) & Country level (13 key countries The U.S, Canada, Germany, France, UK, Italy, China, Japan, India, Middle East, Africa, South America)
Inquire or share your questions, if any: https://i2iresearch.com/report/global-cancer-gene-therapy-market-2020-market-size-share-growth-trends-forecast-2025/
Key questions answered in the report:1. What is the current size of the Cancer Gene Therapy Market, at a global, regional & country level?2. How is the market segmented, who are the key end user segments?3. What are the key drivers, challenges & trends that is likely to impact businesses in the Cancer Gene Therapy Market?4. What is the likely market forecast & how will be Cancer Gene Therapy Market impacted?5. What is the competitive landscape, who are the key players?6. What are some of the recent M&A, PE / VC deals that have happened in the Cancer Gene Therapy Market?
The report also analysis the impact of COVID 19 based on a scenario-based modelling. This provides a clear view of how has COVID impacted the growth cycle & when is the likely recovery of the industry is expected to pre-covid levels.
Contact us:i2iResearch info to intelligenceLocational Office: *India, *United States, *GermanyEmail: [emailprotected]Toll-free: +1-800-419-8865 | Phone: +91 98801 53667
Read the rest here:
Cancer Gene Therapy Market Research Report with Revenue, Gross Margin, Market Share and Future Prospects till 2026 - Cheshire Media
Recommendation and review posted by Bethany Smith
The Stem Cell-Derived Cells market to Scale new heights in the next decade – Khabar South Asia
Stem cell-derived cells are ready-made human induced pluripotent stem cells (iPS) and iPS-derived cell lines that are extracted ethically and have been characterized as per highest industry standards. Stem cell-derived cells iPS cells are derived from the skin fibroblasts from variety of healthy human donors of varying age and gender. These stem cell-derived cells are then commercialized for use with the consent obtained from cell donors. These stem cell-derived cells are then developed using a complete culture system that is an easy-to-use system used for defined iPS-derived cell expansion. Majority of the key players in stem cell-derived cells market are focused on generating high-end quality cardiomyocytes as well as hepatocytes that enables end use facilities to easily obtain ready-made iPSC-derived cells. As the stem cell-derived cells market registers a robust growth due to rapid adoption in stem cellderived cells therapy products, there is a relative need for regulatory guidelines that need to be maintained to assist designing of scientifically comprehensive preclinical studies. The stem cell-derived cells obtained from human induced pluripotent stem cells (iPS) are initially dissociated into a single-cell suspension and later frozen in vials. The commercially available stem cell-derived cell kits contain a vial of stem cell-derived cells, a bottle of thawing base and culture base.
The increasing approval for new stem cell-derived cells by the FDA across the globe is projected to propel stem cell-derived cells market revenue growth over the forecast years. With low entry barriers, a rise in number of companies has been registered that specializes in offering high end quality human tissue for research purpose to obtain human induced pluripotent stem cells (iPS) derived cells. The increase in product commercialization activities for stem cell-derived cells by leading manufacturers such as Takara Bio Inc. With the increasing rise in development of stem cell based therapies, the number of stem cell-derived cells under development or due for FDA approval is anticipated to increase, thereby estimating to be the most prominent factor driving the growth of stem cell-derived cells market. However, high costs associated with the development of stem cell-derived cells using complete culture systems is restraining the revenue growth in stem cell-derived cells market.
To remain ahead of your competitors, request for a sample[emailprotected]
https://www.persistencemarketresearch.com/samples/28780
The global Stem cell-derived cells market is segmented on basis of product type, material type, application type, end user and geographic region:
Segmentation by Product Type
Segmentation by End User
The stem cell-derived cells market is categorized based on product type and end user. Based on product type, the stem cell-derived cells are classified into two major types stem cell-derived cell kits and accessories. Among these stem cell-derived cell kits, stem cell-derived hepatocytes kits are the most preferred stem cell-derived cells product type. On the basis of product type, stem cell-derived cardiomyocytes kits segment is projected to expand its growth at a significant CAGR over the forecast years on the account of more demand from the end use segments. However, the stem cell-derived definitive endoderm cell kits segment is projected to remain the second most lucrative revenue share segment in stem cell-derived cells market. Biotechnology and pharmaceutical companies followed by research and academic institutions is expected to register substantial revenue growth rate during the forecast period.
To receive extensive list of important regions, Request Methodology here @
https://www.persistencemarketresearch.com/methodology/28780
North America and Europe cumulatively are projected to remain most lucrative regions and register significant market revenue share in global stem cell-derived cells market due to the increased patient pool in the regions with increasing adoption for stem cell based therapies. The launch of new stem cell-derived cells kits and accessories on FDA approval for the U.S. market allows North America to capture significant revenue share in stem cell-derived cells market. Asian countries due to strong funding in research and development are entirely focused on production of stem cell-derived cells thereby aiding South Asian and East Asian countries to grow at a robust CAGR over the forecast period.
Some of the major key manufacturers involved in global stem cell-derived cells market are Takara Bio Inc., Viacyte, Inc. and others.
You Can Request for TOC[emailprotected]
https://www.persistencemarketresearch.com/toc/28780
Explore Extensive Coverage of PMR`s
Life Sciences & Transformational HealthLandscape
About us:
Persistence Market Research (PMR) is a third-platform research firm. Our research model is a unique collaboration of data analytics andmarket research methodologyto help businesses achieve optimal performance.
To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes.
Our client success stories feature a range of clients from Fortune 500 companies to fast-growing startups. PMRs collaborative environment is committed to building industry-specific solutions by transforming data from multiple streams into a strategic asset.
Contact us:
Naved BegPersistence Market ResearchAddress 305 Broadway, 7th Floor, New York City,NY 10007 United StatesU.S. Ph. +1-646-568-7751USA-Canada Toll-free +1 800-961-0353Sales[emailprotected]Websitehttps://www.persistencemarketresearch.com
See the original post here:
The Stem Cell-Derived Cells market to Scale new heights in the next decade - Khabar South Asia
Recommendation and review posted by Bethany Smith
The Stem Cell-Derived Cells Market to witness explicit growth from 2019 and 2029 – The Haitian-Caribbean News Network
Stem cell-derived cells are ready-made human induced pluripotent stem cells (iPS) and iPS-derived cell lines that are extracted ethically and have been characterized as per highest industry standards. Stem cell-derived cells iPS cells are derived from the skin fibroblasts from variety of healthy human donors of varying age and gender. These stem cell-derived cells are then commercialized for use with the consent obtained from cell donors. These stem cell-derived cells are then developed using a complete culture system that is an easy-to-use system used for defined iPS-derived cell expansion. Majority of the key players in stem cell-derived cells market are focused on generating high-end quality cardiomyocytes as well as hepatocytes that enables end use facilities to easily obtain ready-made iPSC-derived cells. As the stem cell-derived cells market registers a robust growth due to rapid adoption in stem cellderived cells therapy products, there is a relative need for regulatory guidelines that need to be maintained to assist designing of scientifically comprehensive preclinical studies. The stem cell-derived cells obtained from human induced pluripotent stem cells (iPS) are initially dissociated into a single-cell suspension and later frozen in vials. The commercially available stem cell-derived cell kits contain a vial of stem cell-derived cells, a bottle of thawing base and culture base.
The increasing approval for new stem cell-derived cells by the FDA across the globe is projected to propel stem cell-derived cells market revenue growth over the forecast years. With low entry barriers, a rise in number of companies has been registered that specializes in offering high end quality human tissue for research purpose to obtain human induced pluripotent stem cells (iPS) derived cells. The increase in product commercialization activities for stem cell-derived cells by leading manufacturers such as Takara Bio Inc. With the increasing rise in development of stem cell based therapies, the number of stem cell-derived cells under development or due for FDA approval is anticipated to increase, thereby estimating to be the most prominent factor driving the growth of stem cell-derived cells market. However, high costs associated with the development of stem cell-derived cells using complete culture systems is restraining the revenue growth in stem cell-derived cells market.
To remain ahead of your competitors, request for a sample[emailprotected]
https://www.persistencemarketresearch.com/samples/28780
The global Stem cell-derived cells market is segmented on basis of product type, material type, application type, end user and geographic region:
Segmentation by Product Type
Segmentation by End User
The stem cell-derived cells market is categorized based on product type and end user. Based on product type, the stem cell-derived cells are classified into two major types stem cell-derived cell kits and accessories. Among these stem cell-derived cell kits, stem cell-derived hepatocytes kits are the most preferred stem cell-derived cells product type. On the basis of product type, stem cell-derived cardiomyocytes kits segment is projected to expand its growth at a significant CAGR over the forecast years on the account of more demand from the end use segments. However, the stem cell-derived definitive endoderm cell kits segment is projected to remain the second most lucrative revenue share segment in stem cell-derived cells market. Biotechnology and pharmaceutical companies followed by research and academic institutions is expected to register substantial revenue growth rate during the forecast period.
To receive extensive list of important regions, Request Methodology here @
https://www.persistencemarketresearch.com/methodology/28780
North America and Europe cumulatively are projected to remain most lucrative regions and register significant market revenue share in global stem cell-derived cells market due to the increased patient pool in the regions with increasing adoption for stem cell based therapies. The launch of new stem cell-derived cells kits and accessories on FDA approval for the U.S. market allows North America to capture significant revenue share in stem cell-derived cells market. Asian countries due to strong funding in research and development are entirely focused on production of stem cell-derived cells thereby aiding South Asian and East Asian countries to grow at a robust CAGR over the forecast period.
Some of the major key manufacturers involved in global stem cell-derived cells market are Takara Bio Inc., Viacyte, Inc. and others.
You Can Request for TOC[emailprotected]
https://www.persistencemarketresearch.com/toc/28780
Explore Extensive Coverage of PMR`s
Life Sciences & Transformational HealthLandscape
About us:
Persistence Market Research (PMR) is a third-platform research firm. Our research model is a unique collaboration of data analytics andmarket research methodologyto help businesses achieve optimal performance.
To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes.
Our client success stories feature a range of clients from Fortune 500 companies to fast-growing startups. PMRs collaborative environment is committed to building industry-specific solutions by transforming data from multiple streams into a strategic asset.
Contact us:
Naved BegPersistence Market ResearchAddress 305 Broadway, 7th Floor, New York City,NY 10007 United StatesU.S. Ph. +1-646-568-7751USA-Canada Toll-free +1 800-961-0353Sales[emailprotected]Websitehttps://www.persistencemarketresearch.com
Read more here:
The Stem Cell-Derived Cells Market to witness explicit growth from 2019 and 2029 - The Haitian-Caribbean News Network
Recommendation and review posted by Bethany Smith
The roles bone marrow plays in the body – Therogersvillereview
Bones make up the skeletal system and serve the important function of giving the body support to be able to move. Whats inside the bones also is essential to personal health.
Bone marrow can be found in the center of bones. According to the online wellness resource Healthline, this viscous or spongy tissue comes in two types: red or yellow bone marrow. Both have specific functions in the body.
Red bone marrow is essential for a process called hematopoiesis, or blood cell production. Hematopoietic stem cells in the red bone marrow can develop into key blood cells, including red blood cells, which carry oxygen-rich blood to the body; platelets, which help blood to clot; and white blood cells, which are involved in immune system responses.
Yellow bone marrow is involved with the storage of fats. These fats can be used as an energy source as needed. Yellow bone marrow also contains mesenchymal stem cells that can develop into bone, fat, muscle, or cartilage cells.
Over time, yellow bone marrow replaces red bone marrow in most of the bones in the adult body. Only a few bones, such as the pelvis, skull, vertebrae, and ribs, will contain red bone marrow into adulthood.
According to Medical News Today, bone marrow makes more than 200 billion new blood cells every day. Most blood cells in the body develop from bone marrow cells.
Issues with bone marrow can produce a host of side effects. Fatigue or weakness, fever, increased infections, easy bleeding and bruising, and specific conditions like leukemia and anemia can develop as a result of bone marrow-related problems. In some cases, a bone marrow transplant may be needed to replace diseased or nonfunctioning bone marrow. It also may help regenerate a new immune system that can fight leukemia or other cancers.
Bone marrow transplants also may involve replacing existing bone marrow with genetically healthy bone marrow to prevent future damage from certain genetic diseases, according to Medical News Today. Bone marrow transplants can come from ones own stem cells, a twin, a sibling, parent, or an unrelated donor. Marrow transplants also may come from stored umbilical cord blood.
Bone marrow is vital to the overall health and function of the human body. Bone marrow affects just about every other cell due to its unique relationship with blood production and immune function.
See the original post:
The roles bone marrow plays in the body - Therogersvillereview
Recommendation and review posted by Bethany Smith
Severe Infections Wreak Havoc on Mouse Blood Cell Production – Technology Networks
Severe infections like malaria cause short and long-term damage to precursor blood cells in mice, but some damage could be reversed, find researchers.
A team led by researchers from Imperial College London and The Francis Crick Institute have discovered that severe infections caused by malaria disrupt the processes that form blood cells in mice. This potentially causes long-term damage that could mean people who have recovered from severe infections are vulnerable to new infections or to developing blood cancers.
The team also discovered that the damage could be reduced or partially reversed in mice with a hormone treatment that regulates bone calcium coupled with an antioxidant. The research could lead to new ways of preventing long-term damage from severe infections including malaria, TB and COVID-19.
First author Dr Myriam Haltalli, who completed the work while at the Department of Life Sciences at Imperial, said: "We discovered that malaria infection reprograms the process of blood cell production in mice and significantly affects the function of precursor blood cells. These changes could cause long-term alterations, but we also found a way to significantly reduce the amount of damage and potentially rescue the healthy production of blood cells."
Unexpectedly fast changes
Blood is made up of several different cell types, that all originate as haematopoietic stem cells (HSCs) in the bone marrow. During severe infection, the production of all blood cells ramps up to help the body fight the infection, depleting the HSCs.
Now, the team has shown how infections also damage the bone marrow environment that is crucial for healthy HSC production and function. They discovered this using advanced microscopy technologies at Imperial and the Crick, RNA analyses led by the Gottgens group at Cambridge University, and mathematical modelling led by Professor Ken Duffy at Maynooth University.
The mice developed malaria naturally, following bites from mosquitoes carrying Plasmodium parasites, provided by Dr Andrew Blagborough at Cambridge University. The researchers subsequently observed the changes in the bone marrow environment and the effect on HSC function.
Within days of infection, blood vessels became leaky and there was a dramatic loss in bone-forming cells called osteoblasts. These changes appear strongly linked to the decline in the pool of HSCs during infection.
Lead author Professor Cristina Lo Celso, from the Department of Life Sciences at Imperial, said: "We were surprised at the speed of the changes, which was completely unexpected. We may think of bone as an impenetrable fortress, but the bone marrow environment is incredibly dynamic and susceptible to damage."
Reducing the pool of HSCs can have several consequences. In the short-term, it appears to particularly affect the production of neutrophils - white blood cells that form an essential part of the immune system. This can leave patients vulnerable to further infections, with potentially long-term consequences for the functioning of the immune system.
In the long term, the pool of HSCs may remain below normal levels, which can increase the chances of the patient developing blood cancers like leukaemia.
Mitigating the impacts
After determining in detail how severe infection affects the bone marrow environment and HSC function, the team tested a way to prevent the damage. Before infecting the mice, they treated them with a hormone that regulates bone calcium and an antioxidant to counter cellular oxidative stress, and then again after infection.
This process led to a tenfold increase in HSC function following infection compared to mice that received no treatment (around 20-40 per cent function compared to two percent function, respectively). Although this is not a complete recovery, the vast increase in function is a positive sign.
The team note that the requirement to start the hormone treatment before infection, combined with its expense and need to be refrigerated, make it unviable as a solution, especially in many parts of the world where severe infections like malaria and TB are prevalent.
However, they hope that proof that the impact of severe infection on HSC function can be significantly lessened will lead to the development of new treatments that can be widely administered.
Professor Lo Celso said: "The long-term impacts of COVID-19 infection are just starting to be known. The impact on HSC function appears similar across multiple severe infections, suggesting our work on malaria could shed light on the possible long-term consequences of COVID-19, and how we might mitigate them."
Dr Haltalli concluded: "Protecting HSC function while still developing strong immune responses is key for healthy ageing."
Reference: Haltalli MLR, Watcham S, Wilson NK, et al. Manipulating niche composition limits damage to haematopoietic stem cells during Plasmodium infection. Nat. Cell Biol. 2020:1-12. doi:10.1038/s41556-020-00601-w
This article has been republished from the following materials. Note: material may have been edited for length and content. For further information, please contact the cited source.
Read the original post:
Severe Infections Wreak Havoc on Mouse Blood Cell Production - Technology Networks
Recommendation and review posted by Bethany Smith
Revenue from the Sales of Hematopoietic Stem Cells Transplantation Market to Witness Relatively Significant Growth During 2017 2025 – Canaan Mountain…
Hematopoietic stem cells are young or immature blood cells found to be living in bone marrow. These blood cells on mature in bone marrow and only a small number of these cells get to enter blood stream. These cells that enter blood stream are called as peripheral blood stems cells. Hematopoietic stem cells transplantation is replacement of absent, diseased or damaged hematopoietic stem cells due to chemotherapy or radiation, with healthy hematopoietic stem cells. Over last 30 years hematopoietic stem cells transplantation market seen rapid expansion and constant expansion with lifesaving technological advances. Hematopoietic stem cells transplantation is also known blood and marrow transplantation which brings about reestablishment of the patients immune and medullary function while treating varied range of about 70 hematological and non-hematological disorders. In general hematopoietic stem cells transplantation is used in treatment of hereditary, oncological, immunological and malignant and non-malignant hematological diseases.
There are two types of peripheral blood stem cell transplants mainly autologous and allogeneic transplantation. In autologous transplants patients own hematopoietic stem cells are harvested or removed before the high-dose treatment that might destroy the patients hematopoietic stem cells. While in allogeneic transplants stem cells are obtained from a tissue type of matched or mismatched donor. Hematopoietic stem cells are harvested from blood or bone marrow and is then frozen to use later. Depending upon the source of hematopoietic stem cells, worldwide there are three types of hematopoietic stem cells transplants namely bone marrow transplant (BMT), peripheral blood stem cell transplant and cord blood transplant. Major drivers in the hematopoietic stem cells transplantation market are establishment of strong and well developed network of hematopoietic stem cells transplantation organizations having global reach and presence has recognized NGO named Worldwide Network for Blood and Marrow Transplantation Group (WBMT) in official relation with World Health Organization (WHO) and rapid increase in number of transplants. Major restraints in hematopoietic stem cells transplantation market is high cost of transplantation and lack of funding for WBMT and other organizations such as regional, national and donor.
Get To Know Sample of Report @ https://www.persistencemarketresearch.com/samples/14563
The global market for Hematopoietic stem cells transplantation market is segmented on basis of transplant type, application, disease indication, end user and geography:
Based on transplantation type, hematopoietic stem cells transplantation market is segmented into allogeneic and autologous. Hematopoietic stem cells transplantation market is also segmented by application type into bone marrow transplant (BMT), peripheral blood stem cell transplant and cord blood transplant. The market for hematopoietic stem cells transplantation is majorly driven by bone marrow transplant (BMT) segment. Based on end user hematopoietic stem cells transplantation market is segmented into hospitals and specialty centers. Peripheral blood stem cell transplant type holds the largest market for hematopoietic stem cells transplantation. Hematopoietic stem cells transplantation market is further segmented by disease indication into three main categories i.e. lymphoproliferative disorders, leukemia, and non-malignant disorders. Segment lymphoproliferative disorder holds largest share amongst the three in Hematopoietic stem cells transplantation market. On the basis of regional presence, global hematopoietic stem cells transplantation market is segmented into five key regions viz. North America, Latin America, Europe, Asia Pacific, and Middle East & Africa. Europe leads the global hematopoietic stem cells transplantation market followed by U.S. due to easy technological applications, funding and high income populations. Other reasons for rise in hematopoietic stem cells transplantation market is high prevalence of lymphoproliferative disorders and leukemia; demand for better treatment options; and easy accessibility and acceptance of population to new technological advances. Transplantation rates in high income countries are increasing at a greater extent but continued rise is also seen in low income countries and expected to rise more. Hematopoietic stem cells transplantation market will have its potential in near future as being a perfect alternative to traditional system in many congenital and acquired hematopoietic disorders management. While India, China and Japan will be emerging as potential markets. An excellent and long term alternative to relief by side effects of chemotherapy, radiotherapy and immune-sensitive malignancies is another driver for hematopoietic stem cells transplantation market. The key players in global hematopoietic stem cells transplantation market are Lonza, Escape Therapeutics, Cesca Therapeutics Inc., Regen BioPharma, Inc., Invitrx Inc, StemGenex, Lion Biotechnologies, Inc., CellGenix GmbH, Actinium Pharmaceuticals, Inc., Pluristem, Kite Pharma, Novartis AG.
Request For TOC @ https://www.persistencemarketresearch.com/toc/14563
The report covers exhaustive analysis on:
Regional analysis includes
Report Highlights:
Access Full Report @ https://www.persistencemarketresearch.com/checkout/14563
About us:
Persistence Market Research (PMR) is a third-platform research firm. Our research model is a unique collaboration of data analytics and market research methodology to help businesses achieve optimal performance.
To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes.
Our client success stories feature a range of clients from Fortune 500 companies to fast-growing startups. PMRs collaborative environment is committed to building industry-specific solutions by transforming data from multiple streams into a strategic asset.
Contact us:
Jayprakash Sharma
Persistence Market Research
Address 305 Broadway, 7th FloorNew York City,
NY 10007 United States
U.S. Ph. +1-646-568-7751
USA-Canada Toll-free +1 800-961-0353
Sales [emailprotected]
Website https://www.persistencemarketresearch.com
See the original post:
Revenue from the Sales of Hematopoietic Stem Cells Transplantation Market to Witness Relatively Significant Growth During 2017 2025 - Canaan Mountain...
Recommendation and review posted by Bethany Smith
Approval of Phase I/II Clinical Trial of ATG-016 (Eltanexor), a Second Generation Selective Inhibitor of Nuclear Export (SINE), in Mainland China for…
SHANGHAI and HONG KONG, Nov. 25, 2020 /PRNewswire/ -- Antengene Corporation Limited ("Antengene", HKSE stock code: 6996.HK), a leading innovative biopharmaceutical company dedicated to discovering, developing and commercializing global first-in-class and/or best-inclass therapeutics in hematology and oncology, announced that the National Medical Products Administration (NMPA) has approved the clinical trial of ATG-016 (eltanexor) in patients with intermediate and higher risk myelodysplastic syndrome (MDS) according to the Revised International Prognostic Scoring System (IPSS-R) after the failure of hypomethylating agents (HMA) based therapy. The trial is a Phase I/II, single-arm, open-label clinical study, aiming to evaluate the pharmacokinetics, safety and efficacy of ATG-016 (eltanexor) monotherapy.
MDS is a heterogeneous group of clonal disorders of the bone marrow hematopoietic stem cells (HPSCs), characterized by ineffective hematopoiesis with peripheral blood cytopenia and a higher risk for developing acute myeloid leukemia (AML). Patients with high-risk MDS refractory to hypomethylating agents have a median overall survival (OS) of only 4 to 6 months with limited options for follow-up treatment. Pre-clinical studies have demonstrated that selective inhibitor of nuclear export (SINE) compounds are able to block the nuclear export of many tumor suppressor proteins (e.g. p53, IkB, p21) leading to their accumulation and activation in the nucleus thereby exerting anti-tumor effects. In addition, SINE compounds can also reduce the nuclear export and translation of many oncogenic mRNA (c-Myc, Bcl-2, Bcl-6, cyclin D) which are bound to elF4E and result in selective apoptosis of tumor cells. ATG-016 is a member of the latest-generation of SINE compounds. Compared to the first-generation nuclear export inhibitor, ATG-016 demonstrates minimal blood-brain barrier permeability and a broader therapeutic window. It has shown preliminary anti-cancer activity in high-risk MDS patients.
Dr. Jay Mei, the Founder, Chairman and CEO of Antengene expressed, "The approval of the ATG-016 clinical trial demonstrates the efficient execution of the Antengene R&D team and is also the first clinical trial approval obtained by Antengene in mainland China after its listing." He also mentioned, "Selinexor, the first-generation selective inhibitor of nuclear export, has shown extensive activity against hematological malignancies and solid tumors, and has been approved by the FDA for relapsed/refractory multiple myeloma and diffuse large B-cell lymphoma. As a second-generation orally available SINE compound, ATG-016 can reduce the blood-brain barrier penetration, thereby representing a broader therapeutic window with potentially less adverse events and better drug tolerability."
About ATG-016
ATG-016 (eltanexor) is a second-generation selective inhibitor of nuclear export compound. Compared to the first-generation SINE compound, ATG-016 has lower blood-brain barrier penetration and broader therapeutic window which allows more frequent dosing and a longer period of exposure at higher levels with better tolerability. Therefore, ATG-016 may be used to target a wider range of indications. We plan to conduct phase I/II clinical studies for MDS in China, and plan to further develop ATG-016 for cancers with high prevalence in the Asia-Pacific region (such as KRAS-mutant solid tumors) and virus infection related malignancies (such as nasopharyngeal carcinoma).
About Antengene
Antengene Corporation Limited ("Antengene", SEHK: 6996.HK) is a leading clinical-stage Asia-Pacific biopharmaceutical company focused on innovative oncology medicines. Antengene aims to provide the most advanced anti-cancer drugs to patients in China, the Asia Pacific Region and around the world. Since its establishment, Antengene has built a pipeline of 12 clinical and pre-clinical stage assets, obtained 10 investigational new drug (IND) approvals and has 9 ongoing cross-regional clinical trials in Asia Pacific. At Antengene, we focus on developing drug candidates with novel mechanisms of action (MoAs) and first-in-class/best-in-class potential to address significant unmet medical needs. The vision of Antengene is to "Treat Patients Beyond Borders" through research, development and commercialization of first-in-class/best-in-class therapeutics.
Forward-looking statements
The forward-looking statements made in this article relate only to the events or information as of the date on which the statements are made in this article. Except as required by law, we undertake no obligation to update or revise publicly any forward-looking statements, whether as a result of new information, future events or otherwise, after the date on which the statements are made or to reflect the occurrence of unanticipated events. You should read this article completely and with the understanding that our actual future results or performance may be materially different from what we expect. In this article, statements of, or references to, our intentions or those of any of our Directors or our Company are made as of the date of this article. Any of these intentions may alter in light of future development.
SOURCE Antengene Corporation Limited
Recommendation and review posted by Bethany Smith
Meet ‘Iron Boy’: Hempfield Twp. boy fighting cancer is real superhero – Gettysburg Times
Alex Vucish is a superhero. He has nearly one million Tik Tok views of him dancing as Iron Man, and a comic book artist painted a picture of him with Iron Man and titled it My Iron Boy.
What makes him a real hero, though, is that Alex, the son of Michael and Jessica Vucish of Hempfield Township, is not fighting off imaginary bad guys.
Since May, Alex has been fighting for his life.
Doctors at Childrens Hospital of Pittsburgh started him today on an intravenous drip of his own harvested stem cells. Thats part of the continuing battle against the neuroblastoma that invaded his stomach and impacted adjacent organs. He nearly died when side effects of one treatment landed him on life support.
There were many times that we werent sure if he was going to be with us, his mother said.
But after a series of surgeries, treatments, and chemotherapy, things are looking more promising for the boy who turned three in July.
Alex is the strongest person I know and we are going to fight this with everything we have. We will never give up until the day we can say All Done to cancer and we can all breathe again, Jessica Vucish wrote on his public Facebook page, Alex Strong 2020.
Her sons journey has been followed by people all over the world who send encouragement and prayers. The family credits that with holding them together through the toughest times that a child could ever experience.
His symptoms began earlier this year when he didnt want to eat and his sleep was interrupted with bouts of screaming. Several visits to his pediatrician did not bring any diagnosis nor relief.
Jessica Vucish gave birth to their second child, Dominic, on April 20, and soon after, Alexs condition worsened. He lost weight, his stomach was distended, and he was constantly crying. They finally took him to the emergency department at Excela Westmoreland Hospital on May 9.
They took one x-ray and immediately sent us to Childrens Hospital, said Jessica Vucish, who rode in the ambulance with him.
Because of COVID-19, only one parent was permitted in the hospital, so her husband and his mother, Lisa Jacobelli of Jeannette, stayed back to take care of the baby.
Alex was admitted for a night filled with testing.
I think they did four ultrasounds, blood work and a CT scan, his mom said. I woke up at 7 a.m. it was Mothers Day and one of the nurses told me that the doctors found a mass, and that it was huge. It was the worst day of our lives.
Alex had stage 3 neuroblastoma, a cancer that develops from immature nerve cells and most commonly strikes children under the age of 5. His had grown to about five by seven inches in his stomach.
We were totally not prepared for this, Jessica Vucish said. The oncologist came up with a plan, that ok, were going to fight this. For the first two weeks we really mourned the cancer diagnosis and we mourned what he was going through. By the end of the second week, we were thinking, We can beat this.
Surgery confirmed the diagnosis, then chemotherapy started immediately to shrink the tumor. The parents signed papers acknowledging the possible side effects, including death.
We were taken aback by how severe everything ended up, his mom said. Alexs stomach became more distended after the first two days of treatment. The chemo sent him into a downward spiral. The tumor had soaked up Alexs blood and oozed it out into his abdomen. It was wrapped around his organs and that sent him into liver failure. His heart rate went up to 198 and he started foaming at the mouth.
He was rushed to the intensive care unit and put on life support. His organs were failing and the edema (doctors removed three liters of fluid) was almost collapsing his lungs. He was so sick that despite pandemic restrictions, both parents and his grandparents were called to his bedside to say goodbye.
We were prepared to lose him, Jessica Vucish said. I remember being told that we were lucky to have him for the time that we did, and that we had to let him go.
But Alex wasnt going anywhere. He was upgraded from the ICU two weeks later and soon resumed a total of six rounds of chemo. In all, he had three stem cell harvests, another stay in ICU, four visits to the emergency room, and the latest chemo to destroy his immunity in preparation for a bone marrow transplant. There also were seven surgeries, including a 16-hour operation to remove the tumor after round 4 of chemo. That left him with a triangle shaped scar similar to what Iron Man has on his chest from near fatal injuries.
Alex already loved Iron Man. His mother found the superhero costume for him to wear on Halloween and took a video of him dancing. When Tik Tok star Jennings Brower saw it, he got a video of a group of people dancing and posted it side by side with Alexs video.
When comic book inker Romeo Tanghal of New Jersey heard about Alex, he created a portrait of the boy with Iron Man and donated it to the family. Tanghal has an extensive rsum with DC Comics, Marvel Comics and others, including illustrating Batman, the Green Lantern, Star Trek and Wonder Woman.
The love and response from the community and from people we dont even know has been absolutely humbling, Jessica Vucish said.
That included a parade of emergency vehicles and other well-wishers that went by when he was home for his third birthday in July.
Alex will remain in isolation for weeks, possibly longer, to avoid infection, and his mother is staying with him for the first two weeks. Shes not permitted to leave the room. Shes a photographer and lost many sessions because of the pandemic, and that gives her time to spend with her son.
Her husband is involved in the family business, Stellar Precision Components in Jeannette, and is working from home. Hell spend the next block of time with Alex until his wife returns.
Vucish hopes that Alex wont remember the worst of the times all of the procedures that frightened him and that hell remember only that he survived. Hes now in the NED phase, which stands for No Evidence of Disease. But his parents know that the tumor can recur.
Everything thats happening is scary, his mom said. But you just have to do it one day at a time because the only other choice is to not do anything. We would much rather fight for his life.
Read this article:
Meet 'Iron Boy': Hempfield Twp. boy fighting cancer is real superhero - Gettysburg Times
Recommendation and review posted by Bethany Smith
G1 Therapeutics: Depressed Price Before Catalyst, No Apparent Reason – Seeking Alpha
We were discussing G1 Therapeutics (GTHX) in our TPT member chat yesterday. It has been a depressed stock for over a year now, and even today, despite having a PDUFA date in less than 3 months, the stock shows little sign of improvement. Lead drug candidate trilaciclib is a myelopreservation agent supporting chemo regimens that have shown enough data in phase 2 trials that the FDA has allowed GTHX to proceed to an NDA directly from there without requiring a phase 3 trial. Rintodestrant, their second asset, is in early stages of developing but targeting a larger opportunity in breast cancer and is going to announce a major update on December 9; and yet, the stock shows no sign of improvement.
Their current pipeline looks like this:
Source
I covered trilaciclib a year ago. There is not a lot to be added to that except that their NDA was accepted by the FDA on August 17 with a PDUFA date for February 15, 2021. The NDA was accepted under an accelerated review program, which is why PDUFA is occurring in 6 months instead of the regular 10. The Priority Review is based on positive data from three randomized clinical trials showing robust myelopreservation benefits for the drug.
There are currently no available therapies to protect patients from chemotherapy-induced toxicities before they occur, said Raj Malik, M.D., Chief Medical Officer and Senior Vice President, R&D. If approved, trilaciclib would be the first proactively administered myelopreservation therapy that is intended to make chemotherapy safer and reduce the need for rescue interventions, such as growth factor administrations and blood transfusions.
Trilaciclib also has a Breakthrough Therapy Designation - meaning preliminary clinical evidence for trilaciclib shows a clear advantage over available therapy. In the NDA acceptance letter, the FDA also stated that it is currently not planning to hold an advisory committee meeting for the drug.
While undergoing chemotherapy, many patients experience significant myelosuppression, become fatigued and susceptible to infection, and often require transfusions and growth factor administrations, said Jared Weiss, M.D., Lineberger Comprehensive Cancer Center, University of North Carolina Chapel Hill, NC. Preventing bone marrow damage proactively is an opportunity to improve the quality of life of patients receiving chemotherapy for small cell lung cancer and reduce costly rescue interventions.
Myelosuppression, caused by damage to bone marrow stem cells, occurs due to chemotherapy and can cause symptoms like anemia, neutropenia or thrombocytopenia. In clinical trials, trilaciclib has demonstrated strong reduction of chemotherapy-induced myelosuppression, and patients receiving trilaciclib experienced fewer dose delays/reductions, infections, hospitalizations, and need for rescue therapies compared to patients receiving chemotherapy alone.
Another important development is trilaciclibs expanded access program, which means the company is making the drug available to patients while it undergoes the approval process. The EAP usually means the drug is so beneficial that it is unethical to make patients wait even a few months to get it. This is an important development because it shows the value of trilaciclib.
In June, G1 Therapeutics entered into a 3-year US/Puerto Rico co-promotion agreement with Boehringer Engelheim, which has a lot of experience in oncology asset commercialization. GTHX will book revenue and retain full commercialization rights, and will pay Boehringer a promotional fee based on net sales. G1 will pay a promotion fee of a mid-twenties percentage of net sales in the first year of commercialization, which decreases to a low double-digit/high single-digit percentage in the second and third years of commercialization, respectively.
As for the total addressable market or TAM, over 25,000 people in the U.S. and Puerto Rico are diagnosed with SCLC every year. Approximately 90% of SCLC patients receive first-line chemotherapy treatment, and approximately 60% of those patients receive subsequent second-line chemotherapy treatment. That means, there are 22,500 patients in the 1st line setting who will benefit from trilaciclib, and another 13,500 patients in the 2nd line setting. That is a total of 36,000 patients. From research available last year, cost of chemotherapy treatment for SCLC patients was around $60,000. If we assume a 10% cost for trilaciclib, then we have $6000 per patient. So, every year, they are looking at a TAM of $216mn in this one indication in the US. With Boehringers involvement, we can safely assume a 10% penetration within the first two years of approval, especially given the breakthrough designation. So, that is $22mn from the US; similar figure for Europe, and another such figure from the RoW will give us $60mn in about 3 years, with increasing penetration of the market.
This is a conservative estimate. As I wrote in my earlier coverage:
According to a 2027 estimate by Decision Resources Group, trilaciclib has the potential to benefit a significant number of patients beyond SCLC. There were approximately 1 million chemo treated patients including adjuvant/1L CRC, 1L NSCLC, and adjuvant/1L BC in the U.S., Europe, and Japan in 2018. Chemotherapy treated patients include:
68,000 - ES-SCLC (1st Line - 3rd Line)
68,000 - 1st Line BC
126,000 - 1st Line NSCLC
356,000 - Adjuvant & 1st Line CRC
354,000 - Adjuvant BC
This vast cohort of patients can benefit from trilaciclib in combination with chemotherapy. The company estimates the target market to be at $3bn.
Trilaciclib is a first-in-class drug and doesn't have real competition. It could replace current rescue growth factor support treatments, "including Neulasta (pegfigrastim), Neupogen (filgrastim), Procrit (epoeitin alpha), and Aranesp (darbepoetin alfa)."
Trilaciclib is also being evaluated in other cancers. From their press release:
In a randomized trial of women with metastatic triple-negative breast cancer, preliminary data showed that trilaciclib improved overall survival when administered in combination with chemotherapy compared with chemotherapy alone. The company plans to present final overall survival data from this trial in the fourth quarter of 2020.
On November 18, the company announced that final overall survival data from the mTNBC trial was consistent with the above preliminary findings announced last year, and showed that trilaciclib significantly improved median OS for patients treated with trilaciclib in combination with a chemotherapy regimen of gemcitabine/carboplatin.
They will initiate a pivotal trial in mTNBC in 2021 with OS as the primary endpoint.
Trilaciclib is being evaluated in neoadjuvant breast cancer as part of the I-SPY 2 TRIAL, and the company expects to initiate a Phase 3 trial in patients treated with chemotherapy for colorectal cancer in the fourth quarter of 2020.
In the same press release quoted above, GTHX also announced that it will provide an update on Rintodestrant at the 2020 San Antonio Breast Cancer Symposium (SABCS) to be held virtually on December 9, 2020. Specifically, they said The company will also present updated monotherapy findings from the Phase 1 portion of its ongoing clinical trial of rintodestrant, a potential best-in-class oral selective estrogen receptor degrader (SERD) in development for treatment of ER+, HER2- breast cancer.
It seems they will provide pk/pd and mtd (maximum tolerated dose) data, with probable comments on early signs of efficacy. Primary outcomes are dose limiting toxicity and phase 2 dose determination. secondary outcomes are about efficacy, specifically tumor response with RECIST, and pk/pd. Before competitor fulvestrant went generic last year, it had $541mn annual sales. This indicates the potential for Rintodestrant.
I am providing below the entire abstract on Rintodestrant - there will be three of these but this is the critical one.
Background: Rintodestrant (G1T48) is a potent oral selective estrogen receptor degrader (SERD) that competitively binds to the estrogen receptor (ER) and blocks ER signaling in tumors resistant to other endocrine therapies. Preliminary results from Part 1 dose escalation showed robust target engagement on 18F-fluoroestradiol positron emission tomography (FES-PET), a favorable safety profile, and encouraging antitumor activity in patients with heavily pretreated ER+/HER2- advanced breast cancer (ABC), including those with ESR1 mutations (Dees et al., ESMO 2019 [abstract #3587]). Here, we present updated results from dose escalation and expansion (Parts 1 and 2). Methods: This Phase 1, first-in-human, open-label study evaluated rintodestrant monotherapy in women with ER+/HER2- ABC after progression on endocrine therapy. Part 1 was a 3+3 dose escalation (200- 1000 mg once daily [QD]); Part 2 expanded 600 and 1000 mg QD; and Part 3 was added to assess rintodestrant with palbociclib in patients in earlier lines in the advanced setting. Primary objectives included dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), safety, and recommended Phase 2 dose. Secondary objectives included pharmacokinetics and antitumor activity (RECIST v1.1). Exploratory objectives included pharmacodynamic inhibition of ER target engagement (FES-PET), mutation profiling (cell-free DNA [cfDNA]), and change in ER expression from baseline to on-treatment tumor biopsies. Results: As of May 13, 2020, 67 patients (Part 1: n = 26; Part 2: n = 41) were treated, with a median age of 61 years (range 34-83) and ECOG PS of 0 (49%) or 1 (51%). Median number of prior lines in the advanced setting was 2 (range 0-9), including prior fulvestrant (64%), CDK4/6 inhibitor (69%), mTOR inhibitor (22%), and/or chemotherapy (46%). Median number of prior lines of endocrine therapy in the advanced setting was 2 (range 0-5), with 61% of patients having received 2 lines. Treatment-related adverse events (TRAEs) were reported in 70% of patients. The most common TRAEs in 10% of patients included hot flush (24%), fatigue (21%), nausea (19%), diarrhea (18%), and vomiting (10%), mostly grade 1 or 2. No DLTs were reported and MTD was not reached. Dose reduction due to TRAEs occurred in 1 patient (1%), with elevated transaminases (grade 3 ALT and grade 2 AST) at 600 mg. Serious TRAEs occurred in 2 patients at 1000 mg (grade 5 cerebral hemorrhage in the setting of low molecular weight heparin and grade 2 upper abdominal pain). Two patients (3%) discontinued treatment due to TRAEs. Overall, the frequency of patients with TRAEs at 800 mg was comparable with that at 600 mg (57% vs 63%) and less than that at 1000 mg (81%). Of 67 patients, 16 were on study treatment for 24 weeks and 3 (n = 1 at 600 mg; n = 2 at 1000 mg, including 1 with ESR1 mutation) had a confirmed partial response (clinical benefit rate [CBR]: 28%). FES-PET standard uptake values decreased at week 4 with a mean reduction of 87% (8%) at doses 600 mg. Of 59 patients tested for baseline cfDNA, 41% harbored 1 ESR1 mutation, with a similar CBR in both groups (33% in ESR1 mutant and 29% in ESR1 wild-type). Seven of 9 patients had a decrease in ER immunohistochemistry H-score at both 600 and 1000 mg (median [range]: -27.8% [-33.8%, - 3.4%]), irrespective of ESR1 mutation status. Based on safety, efficacy, and ER degradation, 800 mg was selected as the optimal dose for further study. Conclusions: Rintodestrant continues to demonstrate an excellent safety/tolerability profile across all doses, with promising antitumor activity in patients with heavily pretreated ER+/HER2- ABC, including those with tumors harboring ESR1 mutations. Part 3 of this study, evaluating rintodestrant 800 mg QD with palbociclib in a more endocrine-sensitive population, is ongoing (NCT03455270).
There is not a lot to comment here. 800 mg seems to be the optimal dose, and that one patient with grade 5 cerebral hemorrhaging may be an outlier, especially with heparin in the mix, but that is something to keep an eye out for. Effect on an endocrine-sensitive population is preliminary but encouraging, and although a partial response, at this late stage in the game, that is not a bad deal. A larger trial developing the ESR1 scenario more fully is going to be interesting to watch.
According to latest reports, the company has $200-205mn in cash and cash equivalents as of the latest quarter. Burn was around $90mn last year, so with the Boehringer deal, that is not going to increase substantially, putting them in a comfortable position with a 2-year cash runway. The company recently changed CEOs, which seems to be in anticipation of approval, and nothing else.
For whatever reasons I cannot fathom, the stock remains depressed. They have a major catalyst 3 months down the line, and if past performance in trials is anything to go by, theres considerable chance of approval. Post approval scenario, both in terms of revenue stream and label expansions/other indications, looks good. I think this is a good candidate to accumulate at this time.
Thanks for reading. At the Total Pharma Tracker, we offer the following:-
Our Android app and website features a set of tools for DIY investors, including a work-in-progress software where you can enter any ticker and get extensive curated research material.
For investors requiring hands-on support, our in-house experts go through our tools and find the best investible stocks, complete with buy/sell strategies and alerts.
Sign up now for our free trial, request access to our tools, and find out, at no cost to you, what we can do for you.
Disclosure: I/we have no positions in any stocks mentioned, and no plans to initiate any positions within the next 72 hours. I wrote this article myself, and it expresses my own opinions. I am not receiving compensation for it (other than from Seeking Alpha). I have no business relationship with any company whose stock is mentioned in this article.
Read more:
G1 Therapeutics: Depressed Price Before Catalyst, No Apparent Reason - Seeking Alpha
Recommendation and review posted by Bethany Smith
Impact of COVID 19 on Orthopedic Regenerative Medicine Market Detailed Research Study 2020-2027 | Curasan, Inc., Carmell Therapeutics Corporation,…
Orthopedic Regenerative Medicine Market
Coherent Market Insights, 26-11-2020: The research report on the Orthopedic Regenerative Medicine Market is a deep analysis of the market. This is a latest report, covering the current COVID-19 impact on the market. The pandemic of Coronavirus (COVID-19) has affected every aspect of life globally. This has brought along several changes in market conditions. The rapidly changing market scenario and initial and future assessment of the impact is covered in the report. Experts have studied the historical data and compared it with the changing market situations. The report covers all the necessary information required by new entrants as well as the existing players to gain deeper insight.
Furthermore, the statistical survey in the report focuses on product specifications, costs, production capacities, marketing channels, and market players. Upstream raw materials, downstream demand analysis, and a list of end-user industries have been studied systematically, along with the suppliers in this market. The product flow and distribution channel have also been presented in this research report.
Get a sample pages of this Report:https://www.coherentmarketinsights.com/insight/request-sample/3566
The segments and sub-section of Orthopedic Regenerative Medicine market are shown below:
By Procedure Cell TherapyTissue EngineeringBy Cell TypeInduced Pluripotent Stem Cells (iPSCs)Adult Stem CellsTissue Specific Progenitor Stem Cells (TSPSCs),Mesenchymal Stem Cells (MSCs)Umbilical Cord Stem Cells (UCSCs)Bone Marrow Stem Cells (BMSCs)By SourceBone MarrowUmbilical Cord BloodAdipose TissueAllograftsAmniotic FluidBy ApplicationsTendons RepairCartilage RepairBone RepairLigament RepairSpine RepairOthers
Some of the key players/Manufacturers involved in the Orthopedic Regenerative Medicine Market are Curasan, Inc., Carmell Therapeutics Corporation, Anika Therapeutics, Inc., Conatus Pharmaceuticals Inc., Histogen Inc., Royal Biologics, Ortho Regenerative Technologies, Inc., Swiss Biomed Orthopaedics AG, Osiris Therapeutics, Inc., and Octane Medical Inc.
If opting for the Global version of Orthopedic Regenerative Medicine Market analysis is provided for major regions as follows:
North America (The US, Canada, and Mexico)
Europe (the UK, Germany, France, and Rest of Europe)
Asia Pacific (China, India, and Rest of Asia Pacific)
Latin America (Brazil and Rest of Latin America)
Middle East & Africa (Saudi Arabia, the UAE, South Africa, and Rest of Middle East & Africa)
Buy This Complete A Business Report @:https://www.coherentmarketinsights.com/insight/buy-now/3566
The Orthopedic Regenerative Medicine Market Report Consists of the Following Points:
The report consists of an overall prospect of the market that helps gain significant insights about the global market.
The market has been categorized based on types, applications, and regions. For an in-depth analysis and better understanding of the market, the key segments have been further categorized into sub-segments.
The factors responsible for the growth of the market have been mentioned. This data has been gathered from primary and secondary sources by industry professionals. This provides an in-depth understanding of key segments and their future prospects.
The report analyses the latest developments and the profiles of the leading competitors in the market.
The Orthopedic Regenerative Medicine Market research report offers an eight-year forecast.
Enquire for customization in Report @https://www.coherentmarketinsights.com/insight/request-customization/3566
Interested about who is winning the race of COVID-19 Vaccine. Coherent Market Insights (CMI)
providesCOVID-19 Vaccine Trackerfor all the latest updates about COVID-19 Vaccine.
CMI also offers Custom Research services providing focused, comprehensive and tailored research according to clientele objectives. Thanks for reading this article; you can also get individual chapter wise section or region wise report like North America, Europe or Asia.
About Us:
Coherent Market Insights is a prominent market research and consulting firm offering action-ready syndicated research reports, custom market analysis, consulting services, and competitive analysis through various recommendations related to emerging market trends, technologies, and potential absolute dollar opportunity.
Contacts Us:Direct Line: +1-206-701-6702 (US)Web:https://www.coherentmarketinsights.com
Recommendation and review posted by Bethany Smith
The FDA has approved the first drug to treat the rapid-aging disease progeria – Science News
The U.S. Food and Drug Administration has approved a treatment that could give children with a rare genetic illness that causes premature aging more time to live.
Children with the disease, known as Hutchinson-Gilford progeria syndrome, or progeria for short, often die of heart failure, heart attack or stroke as teenagers. Most children with the disorder die before they reach age 15. The newly approved drug, called Zokinvy, is the first and only approved treatment for progeria and certain related syndromes, the FDA announced November 20.
In clinical trials of 62 children receiving the drug, Zokinvy increased life span by about 3 months on average during the first three years of treatment, compared with another 81 kids who did not take the drug from a separate study that collected their health data. Following children who continued to receive Zokinvy for up to 11 years showed that, on average, kids life spans were lengthened by about 2.5 years.
This is not a cure, cautions Monica Kleinman, a pediatric critical care doctor at Boston Childrens Hospital who was involved with the clinical trials. Weve hopefully extended the life span that [the children] have by slowing the pace of the disease, but, she says, the drug doesnt give kids a normal length of life.
Headlines and summaries of the latest Science News articles, delivered to your inbox
An estimated 350 to 400 kids across the world have progeria. For these children, a single mutation in their genetic code upends their health (SN: 2/7/13). That mutation interferes with the gene responsible for making the protein lamin A, which helps hold cells nuclei together. Children with progeria end up with higher amounts of a defective protein called progerin, which is similar to lamin A but with an extra piece attached. This protein gets stuck in cells membranes and cant be recycled for fresh proteins, causing the cells to prematurely age and making blood vessels and connective tissue stiffer, Kleinman says.
Everyone makes some progerin, and the body makes more as it gets older, Kleinman explains, but children with progeria make a huge amount. Children typically appear normal at birth, but start to show signs of the illness in their first two years of life. Over their lives, these kids experience loss of hair and body fat, joint stiffness, cardiovascular disease and other symptoms of accelerated aging.
Zokinvy, made by the company Eiger BioPharmaceuticals of Palo Alto, Calif., blocks some of that progerin production, lowering the amount that accumulates in kids cells. But the oral drug, taken as capsules, doesnt fully block production, she says, and the amount that patients can receive is limited by the drugs side effects, which include vomiting, diarrhea and fatigue.
The drug is a testament to the power of basic research, says Tom Misteli, a cell biologist at the National Cancer Institute in Bethesda, Md, who was not involved with work on the drug. Zokinvy builds on decades of research on many aspects of the lamin A protein, including the seemingly esoteric chemical modification that forms progerin, he says.
Nobody studying this protein or the modification could have expected it to become a drug target, Misteli adds. But once the disease-causing gene was identified, researchers zeroed in on the class of drugs that includes Zokinvy as potential treatments.
With the new drug approval, the focus is now to test additional drugs or therapeutics in combination with Zokinvy, Misteli says. That could help lengthen the lives of children with progeria even further. Researchers are also investigating gene therapy approaches, with the goal of fixing the mutation that causes the debilitating illness.
Continued here:
The FDA has approved the first drug to treat the rapid-aging disease progeria - Science News
Recommendation and review posted by Bethany Smith
Stem Cells Market Research Provides an In-Depth Analysis on the Future Growth Prospects and Industry Trends Adopted by the Competitors | (2020-2027),…
Stem Cells Market Overview:
Reports and Data has recently published a new research study titled Global Stem Cells Market that offers accurate insights for the Stem Cells market formulated with extensive research. The report explores the shifting focus observed in the market to offer the readers data and enable them to capitalize on market development. The report explores the essential industry data and generates a comprehensive document covering key geographies, technology developments, product types, applications, business verticals, sales network and distribution channels, and other key segments.
The global Stem Cells market is forecasted to grow at a rate of 8.4% from USD 9.35 billion in 2019 to USD 17.78 billion in 2027.
The report is further furnished with the latest market changes and trends owing to the global COVID-19 crisis. The report explores the impact of the crisis on the market and offers a comprehensive overview of the segments and sub-segments affected by the crisis. The study covers the present and future impact of the pandemic on the overall growth of the industry.
Get a sample of the report @ https://www.reportsanddata.com/sample-enquiry-form/2981
Competitive Landscape:
The global Stem Cells market is consolidated owing to the existence of domestic and international manufacturers and vendors in the market. The prominent players of the key geographies are undertaking several business initiatives to gain a robust footing in the industry. These strategies include mergers and acquisitions, product launches, joint ventures, collaborations, partnerships, agreements, and government deals. These strategies assist them in carrying out product developments and technological advancements.
The report covers extensive analysis of the key market players in the market, along with their business overview, expansion plans, and strategies. The key players studied in the report include:
Thermo Fisher Scientific, Agilent Technologies, Illumina, Inc., Qiagen, Oxford Nanopore Technologies, Eurofins Scientific, F. Hoffmann-La Roche, Danaher Corporation, Bio-Rad Laboratories, and GE Healthcare.
An extensive analysis of the market dynamics, including a study of drivers, constraints, opportunities, risks, limitations, and threats have been studied in the report. The report offers region-centric data and analysis of the micro and macro-economic factors affecting the growth of the overall Stem Cells market. The report offers a comprehensive assessment of the growth prospects, market trends, revenue generation, product launches, and other strategic business initiatives to assist the readers in formulating smart investment and business strategies.
To read more about the report, visit @ https://www.reportsanddata.com/report-detail/stem-cells-market
Product Outlook (Revenue, USD Billion; 2017-2027)
Technology Outlook (Revenue, USD Billion; 2017-2027)
Therapy Outlook (Revenue, USD Billion; 2017-2027)
Application Outlook (Revenue, USD Billion; 2017-2027)
Request a discount on the report @ https://www.reportsanddata.com/discount-enquiry-form/2981
Key Coverage in the Stem Cells Market Report:
Thank you for reading our report. Please get in touch with us if you have any queries regarding the report or its customization. Our team will make sure the report is tailored to meet your requirements.
Take a look at other reports from Reports and Data on PR Newswire:
Hydroxycitronellal Market: Hydroxycitronellal Market To Reach USD 192.7 Million By 2027
Sterile Filtration Market: Sterile Filtration Market To Reach USD 8.48 Billion By 2027 | CAGR: 7.7%
Tissue Diagnostics Market: Tissue Diagnostics Market To Reach USD 5.02 Billion By 2027
UV-C Robot Market: UV-C Robot Market to Reach USD 1.46 Billion by 2027
About Us:
Our in-house experts assist our clients with advice based on their proficiency in the market that helps them in creating a compendious database for the clients. Our team offers expert insights to clients to guide them through their business ventures. We put in rigorous efforts to keep our clientele satisfied and focus on fulfilling their demands to make sure that the end-product is what they desire. We excel in diverse fields of the market and with our services extending to competitive analysis, research and development analysis, and demand estimation among others, we can help you invest your funds in the most beneficial areas for research and development. You can rely on us to provide every significant detail you might need in your efforts to make your business flourish.
Contact Us:
John Watson
Head of Business Development
Reports and Data|Web:www.reportsanddata.com
Direct Line:+1-212-710-1370
E-mail:[emailprotected]
Connect with us:Blogs|LinkedIn|Twitter
Recommendation and review posted by Bethany Smith
Stem Cells Market 2020: Rising with Immense Development Trends across the Globe by 2027 – The Market Feed
Stem Cells Market Overview:
Reports and Data has recently published a new research study titled Global Stem Cells Market that offers accurate insights for the Stem Cells market formulated with extensive research. The report explores the shifting focus observed in the market to offer the readers data and enable them to capitalize on market development. The report explores the essential industry data and generates a comprehensive document covering key geographies, technology developments, product types, applications, business verticals, sales network and distribution channels, and other key segments.
The report is further furnished with the latest market changes and trends owing to the global COVID-19 crisis. The report explores the impact of the crisis on the market and offers a comprehensive overview of the segments and sub-segments affected by the crisis. The study covers the present and future impact of the pandemic on the overall growth of the industry.
Get a sample of the report @ https://www.reportsanddata.com/sample-enquiry-form/2981
Competitive Landscape:
The global Stem Cells market is consolidated owing to the existence of domestic and international manufacturers and vendors in the market. The prominent players of the key geographies are undertaking several business initiatives to gain a robust footing in the industry. These strategies include mergers and acquisitions, product launches, joint ventures, collaborations, partnerships, agreements, and government deals. These strategies assist them in carrying out product developments and technological advancements.
The report covers extensive analysis of the key market players in the market, along with their business overview, expansion plans, and strategies. The key players studied in the report include:
Celgene Corporation, ReNeuron Group plc, Virgin Health Bank, Biovault Family, Mesoblast Ltd., Caladrius, Opexa Therapeutics, Inc., Precious Cells International Ltd., Pluristem, and Neuralstem, Inc., among others.
An extensive analysis of the market dynamics, including a study of drivers, constraints, opportunities, risks, limitations, and threats have been studied in the report. The report offers region-centric data and analysis of the micro and macro-economic factors affecting the growth of the overall Stem Cells market. The report offers a comprehensive assessment of the growth prospects, market trends, revenue generation, product launches, and other strategic business initiatives to assist the readers in formulating smart investment and business strategies.
To read more about the report, visit @ https://www.reportsanddata.com/report-detail/stem-cells-market
Product Outlook (Revenue, USD Billion; 2017-2027)
Technology Outlook (Revenue, USD Billion; 2017-2027)
Therapy Outlook (Revenue, USD Billion; 2017-2027)
Application Outlook (Revenue, USD Billion; 2017-2027)
Request a discount on the report @ https://www.reportsanddata.com/discount-enquiry-form/2981
Key Coverage in the Stem Cells Market Report:
Thank you for reading our report. Please get in touch with us if you have any queries regarding the report or its customization. Our team will make sure the report is tailored to meet your requirements.
Take a look at other reports from Reports and Data on PR Newswire:
Hydroxycitronellal Market: Hydroxycitronellal Market To Reach USD 192.7 Million By 2027
Sterile Filtration Market: Sterile Filtration Market To Reach USD 8.48 Billion By 2027 | CAGR: 7.7%
Tissue Diagnostics Market: Tissue Diagnostics Market To Reach USD 5.02 Billion By 2027
UV-C Robot Market: UV-C Robot Market to Reach USD 1.46 Billion by 2027
About Us:
Our in-house experts assist our clients with advice based on their proficiency in the market that helps them in creating a compendious database for the clients. Our team offers expert insights to clients to guide them through their business ventures. We put in rigorous efforts to keep our clientele satisfied and focus on fulfilling their demands to make sure that the end-product is what they desire. We excel in diverse fields of the market and with our services extending to competitive analysis, research and development analysis, and demand estimation among others, we can help you invest your funds in the most beneficial areas for research and development. You can rely on us to provide every significant detail you might need in your efforts to make your business flourish.
Contact Us:
John Watson
Head of Business Development
Reports and Data|Web:www.reportsanddata.com
Direct Line:+1-212-710-1370
E-mail:[emailprotected]
Connect with us:Blogs|LinkedIn|Twitter
Originally posted here:
Stem Cells Market 2020: Rising with Immense Development Trends across the Globe by 2027 - The Market Feed
Recommendation and review posted by Bethany Smith
U.S. engineering firms ride on waves of innovation – Building Design + Construction
Last year, SSOE Group saved an automotive client more than $250,000 by streamlining how data from the clients manufacturing structures were gathered, using laser scanning, innovative workflows, and technology that converts point clouds to Revit models that can be imported to structural analysis software.
Innovation continues to be engineering firms best foot forward to remain competitive and relevant. And sometimes, innovation is resolutely basic: For a series of manhole inspections it performed for Carnegie Mellon University, Wiley|Wilson attached a camera controlled by a smartphone app to a 9-foot-long selfie stick for 360-degree information capture. KCI Technologies piloted AM Gradiometry technology, a subsurface investigation methodology that harnesses the power of AM-band radio to identify and map underground infrastructure and anomalies.
Admittedly, most other innovations engineers came up with werent as rudimentary. Jensen Hughes launched a new software program, HazAdvisr, that quickly categorizes chemical hazards and applies them to a project to achieve compliance, eliminating the need for time-consuming and often error-prone classification done by hand. AECOMs patented water treatment solution, De-Fluoro, destroys a globally pervasive emerging contaminant Perfluorooctanoic Acid (PFAS), and optimizes infrastructure upgrades.
Thornton Tomasetti launched Beacon, an embodied carbon measurement tool that allows structural engineers to understand and manage embodied carbon optimization. And CDM Smith recently collaborated with Autodesk to develop the Rapid Energy Modeling tool, an integrated desktop application that enables energy managers and planners to conduct energy analyses at facilities without deploying physical resources onsite. Syska Hennessy was of the same mindset when it established a process to perform remote commissioning, punch-lists, and onsite field work virtually.
Engineers are more frequently being called upon for solutions that reduce their customers risks. For example, Affiliated Engineers Inc.s resilience planning and design service features a tool that addresses climate change for the owners project location, characterizes the risk of failure to engineering systems should the identified potential disaster scale hazardous event(s) occur, develops adaptation and mitigation strategies, and presents this information in rich graphic form to the owner and design team.
Other innovations are designed as platforms for collaboration and greater efficiency. KLH Engineers created a series of custom Revit add-ins geared toward eliminating repetitive, rules-based tasks and providing the engineers with information they need to make informed decisions early in a project. And PBS Engineers is using 3D cameras to document the existing conditions of spaces during the initial site surveys. This process allows the team to have an accurate representation of the MEP conditions for a more coordinated design set.
These innovations emerged at a time when engineering firms were adding to their practice menus, and the sector continued to consolidate. Last October, IPSIntegrated launched a new service called CarTon, a complete cell and gene therapy operational readiness solution. This bundled service offering focuses on getting compliant cell and gene therapy products to market. Bernhards Energy-as-a-Service solutions offer healthcare clients alternative financing and project delivery methods that are designed to reduce cost structure and increase operational margins.
Burns & McDonnell hired a dozen professionals to expand services in the life sciences industry, collectively adding more than 200 years of additional design and construction experience in the pharmaceutical, biotech, animal health, medical device, and gene therapy sectors. EAPC Architects is now offering Entitlement services that process land development cases for rezoning properties, obtaining special permits and conditional uses for a specific land use. HPE Data Center relaunched its consulting practice in the U.S., Thailand, and Indonesia. Davis, Bowen & Friedel introduced the addition of in-house unmanned aerial vehicle (UAV) drone services. And DeSimone Consulting Engineers initiated a Risk Management Services practice.
For its industrializing colocation and hyperscale clientele, EYP Mission Critical rolled out a suite of formalized services focused on the adaptation of standardized designs for new paradigms, modular containerized implementation, tenant fit-out/retrofit, and optimization and operational efficiency in power, space, and cooling.
The coronavirus pushed several firms into new areas. Milhouse Engineerings environmental experts developed a method for sanitization that combines a specially atomized fog and a three-stage HEPA air filtration system. NV5 Global offered a suite of COVID-19 support services. To support business continuity, its COVID-19 facility health and safety services provide site-specific deep cleaning protocols and training of cleaning staff to minimize risk of exposures. In California, the firm offered third-party building inspections and plan reviews in municipalities that closed their building departments during the outbreak. NV5s MEP engineering and commissioning group supports remote field hospitals and facility renovations.
NV5 Global made nine strategic acquisitions in 2019 that added 1,100 employees, enhanced service offerings in the environmental, technology, infrastructure, and energy markets; and broadened its geographic coverage.
In 2019, IMEG Corp. acquired five firms and 10 new office locations. These acquisitions brought its employee count to 1,500 employees across nearly 50 locations. In 2019, Dewberry acquired California-based Drake Haglan and Associates, an 80-person firm serving private- and public-sector clients. P2S Inc. acquired Muni-Fed, an energy and civil engineering consulting firm, in Q4 2019. In August 2019, TLC Engineering acquired an FP/LS firm that doubled TLCs revenue in this discipline. It also acquired a Chicago-based MEP firm that became its 15th location. And with the addition of design firms studio951 and EPOCH during the fourth quarter of 2019, Shive-Hattery expanded into two new markets: Lincoln, Neb., and South Bend, Ind.
Other firms saw operational opportunities in the virtual world: For instance, Ross & Baruzzini converted its entire IT infrastructure to Amazon Web Services cloud and virtual desktop services.
Last year, 31% of Desimones revenue came from green building and sustainability projects. And it was easy to forget, during a pandemic, that carbon neutrality remains a long-range goal for the built environment.
Morrison Hershfield, in collaboration with Humber College and project partners, completed a holistic deep energy retrofit of an aging Humber Building NX, making it the first existing building retrofit in Canada to achieve Zero Carbon Building-Design Certification from the Canada Green Building Council. In Spokane, Wash., McKinstry developed and designed Catalyst, a five-story, 159,000-sf cross-laminated timber (CLT) building whose goal is to be one of the largest zero-energy and one of the first zero-carbon buildings in North America.
Read the original:
U.S. engineering firms ride on waves of innovation - Building Design + Construction
Recommendation and review posted by Bethany Smith
Flight Tests to Show B61-12 Will Work on Air Force’s Newest Fighter Jet – I-Connect007
A mock B61-12s strike in the dusty Nevada desert successfully completed the first in a series of flight tests with the U.S. Air Forces newest fighter jet, demonstrating the bombs first release from an internal bomb bay at greater than the speed of sound.
The flight test of the B61-12 with theF-35A Lightning IIthis summer was the first ever at Sandia National Laboratories Tonopah Test Range featuring the fighter jet. It was also the first of a testing series that will conclude with full-weapon systems demonstrations designed to increase confidence the bomb will always work when needed and never under any other circumstances.
Were showing the B61-12s larger compatibility and broader versatility for the countrys nuclear deterrent, and were doing it in the world of COVID-19, said Steven Samuels, a manager with Sandias B61-12 Systems Team. Were not slowing down. Were still moving forward with the B61-12 compatibility activities on different platforms.
In partnership with the National Nuclear Security Administration, Los Alamos National Laboratory and the Air Force, Sandia completed a B61-12full-weapon system demonstrationwith theF-15E Strike Eaglefighter jet in March, and another in July with the Air ForcesB-2 Spiritbomber.
Sandia is the design and engineering lab for non-nuclear components of the nations nuclear stockpile, including the B61-12. In addition to non-nuclear component development, Sandia serves as the technical integrator for the complete weapon, assuring the system meets requirements as a full-weapon system.
Showing the bombs real-world capabilityDuring the Aug. 25 flight test, an F-35A flying faster than the speed of sound dropped a B61-12 containing non-nuclear and mock nuclear components from about 10,500 feet above Tonopah Test Range. The inert B61-12 struck the desert floor in the designated target area about 42 seconds later.
We successfully executed this historic, first-ever F-35A flight test at Tonopah Test Range within the specified delivery criteria, said Brian Adkins, range manager at TTR.
The success of this test, as with all other weapons evaluations, is only possible through the detailed planning, combined with full collaboration between TTR and the program engineers, and the execution of the test evolution by the field operators and recovery specialists in the combined team of Sandia and TTRs operations and maintenance subcontractor, Navarro Research and Engineering, he said.With the multiple phases and operational activities a test involves, the team at TTR is diligent to integrate safety and security into all segments to ensure proper precautions are implemented for mission success.
Coordination between Sandia, Los Alamos, the NNSA and the Air Force made the flight test possible, and initial data shows that all systems and interfaces between the refurbished bomb and the F-35A worked as expected.
Unlike previous fighter jets, the F-35A carries the bomb internally. The recent flight test was the first demonstration of a fully instrumented B61-12 release from an internal bomb bay on a fighter and the first such release at speeds of Mach 1 or greater, Samuels said.
This was the first test to exercise all systems, including mechanical, electrical, communication and release between the B61-12 and the F-35A, he said.
The test also came amidst now commonplace COVID-19 workplace restrictions, which can make planning more difficult but are not slowing down Sandias important mission work, said B61-12 program senior manager Christine Mitchell. Sandia National Labs, Los Alamos National Laboratory, NNSA and our Air Force partners are working diligently to ensure F-35A major milestones stay on track, despite the challenges presented by COVID-19.
The F-35A is a fifth generation fighter and is described by an Air Forceonline fact sheetas an agile, versatile, high-performance, 9-G capable multirole fighter with stealth technology and advanced sensors. Nine countries the United States, the United Kingdom, Italy, Netherlands, Turkey, Canada, Denmark, Norway and Australia were involved in the fighter jets development.
The latest test is a critical piece in the F-35A and B61-12 program, Samuels said. Aboard the newest fighter, the B61-12 provides a strong piece of the overall nuclear deterrence strategy for our country and our allies.
Sandia design and engineering is integral to B61-12 Life Extension ProgramThe compatibility testing is an essential part of theB61-12 Life Extension Programto refurbish, reuse or replace components, extend the bombs service life, and improve its safety, security and effectiveness.
A life extension program allows scientists and engineers to address the aging of nuclear weapons components. Some components are requalified and go back into a weapon without change; others that have aged are remanufactured using the original specifications; and sometimes the original technology is no longer available, so Sandia redesigns those parts using modern technology.
The first B61 entered service 50 years ago, and over the decades numerous modifications have been made to increase safety and reliability. The B61-12 consolidates and replaces most of the previous variants. The National Nuclear Security Administration recentlyannouncedplans to manufacture the first refurbished B61-12 in fiscal year 2022.
Read more:
Flight Tests to Show B61-12 Will Work on Air Force's Newest Fighter Jet - I-Connect007
Recommendation and review posted by Bethany Smith
An F-35A Dropped An Inert B61-12 Nuclear Bomb During Supersonic Flight For The First Time – The Aviationist
An F-35A Lightning II opens its bomb bay doors and drops a mock B61-12 at Sandia National Laboratories Tonopah Test Range. (Screenshot from Sandia National Laboratories video)
Just a few months after the first photos of the F-35 testing the B61-12 nuclear bomb in 2019 for the F-35A Dual Capable Aircraft (DCA) program were released, the Sandia National Laboratories announced that the F-35 dropped for the first time an inert B61-12 nuclear bomb from its internal bomb bay during supersonic flight. According to the press release, the test took place on Aug. 25, 2020 at about 10,500 ft above the Tonopah Test Range, with the inert bomb hitting the target area after a 42 seconds-flight.
The test was the first performed by Sandia, the National Nuclear Security Administration, the Los Alamos National Laboratory and the U.S. Air Force over the Tonopah Test Range with the F-35A and the first of a testing series that will conclude with full-weapon systems demonstrations designed to increase confidence the bomb will always work when needed and never under any other circumstances. Sandia oversees the design and engineering for the non-nuclear components of the United States nuclear stockpile, including the new B61-12, and of the integration of the complete weapon on the aircraft.
We successfully executed this historic, first-ever F-35A flight test at Tonopah Test Range within the specified delivery criteria, said Brian Adkins, range manager at TTR. The success of this test, as with all other weapons evaluations, is only possible through the detailed planning, combined with full collaboration between TTR and the program engineers, and the execution of the test evolution by the field operators and recovery specialists in the combined team of Sandia and TTRs operations and maintenance subcontractor, Navarro Research and Engineering. With the multiple phases and operational activities a test involves, the team at TTR is diligent to integrate safety and security into all segments to ensure proper precautions are implemented for mission success.
Earlier this year, the team completed a B61-12 full-weapon system demonstration with the F-15E Strike Eagle and the B-2A Spirit, completing the integration of the bomb aboard the two aircraft. Differently than the Strike Eagle, the F-35 transports its B61-12 internally, and according to Sandias previous press releases, it seems that the F-15E did not attempt a supersonic drop of the bomb, simply stating that the aircraft was flying near Mach 1 in at least two tests at 1000 ft and 25000 ft. Steven Samuels, a manager with Sandias B61-12 Systems Team, confirmed that the F-35 test was the first demonstration of a fully instrumented B61-12 release from an internal bomb bay on a fighter and the first such release at speeds of Mach 1 or greater.
The F-35A DCA, according to public information, should achieve the nuclear certification in January 2023, pretty much in line with earlier reports that scheduled the completion of the integration works between 2020 and 2022. Regarding the delivery of the new weapon, the B61-12 could be first delivered to the U.S. Air Force starting from 2022, after significant delays were caused by some faulty electrical components. Around 480 bombs will reportedly be upgraded to the new configuration.
As already explained in a previous article here at The Aviationist:
The B61-12 represent the latest LEP (Life-Extention Program) upgrade to the B61 line of nuclear weapons that has already been extensively tested with theF-15E Strike Eaglesof the 422nd Test and Evaluation Squadron,back in 2015.
The Life Extension Program or LEP, will replace the B61 -3, -4, -7, and -10 mods, with the -12 that, along with the B83, will become the only remaining gravity delivered nukes in the inventory.
The B61-12 gravity bomb ensures the current capability for the air-delivered leg of the U.S. strategic nuclear triad well into the future for both bombers and dual-capable aircraft supporting NATO, said Paul Waugh, AFNWCs Air-Delivered Capabilities director in a U.S. Air Forcerelease dated Apr. 13(more or less when the world learnedabout the first use of the famous MOAB in Afghanistan). The B61-12 will be compatible with the B-2A, B-21, F-15E, F-16C/D, F-16 MLU, F-35 and PA-200 aircraft.
The LEP increases the B61s accuracy so much that it will have the same capability against hardened targets as the much more powerful weapons it is replacing.
Here below you can find the video released by Sandia. After the bomb drop at the beginning of the video, you can also see, at about the 30 seconds mark, the spin rocket motors firing to improve stability during the bombs descent. If you cant see the embedded video below, heres the link to the declassified footage.
Originally posted here:
An F-35A Dropped An Inert B61-12 Nuclear Bomb During Supersonic Flight For The First Time - The Aviationist
Recommendation and review posted by Bethany Smith
Flight tests to show B61-12 will work on Air Forces newest fighter jet – Newswise
Newswise ALBUQUERQUE, N.M. A mock B61-12s strike in the dusty Nevada desert successfully completed the first in a series of flight tests with the U.S. Air Forces newest fighter jet, demonstrating the bombs first release from an internal bomb bay at greater than the speed of sound.
The flight test of the B61-12 with the F-35A Lightning II this summer was the first ever at Sandia National Laboratories Tonopah Test Range featuring the fighter jet. It was also the first of a testing series that will conclude with full-weapon systems demonstrations designed to increase confidence the bomb will always work when needed and never under any other circumstances.
Were showing the B61-12s larger compatibility and broader versatility for the countrys nuclear deterrent, and were doing it in the world of COVID-19, said Steven Samuels, a manager with Sandias B61-12 Systems Team. Were not slowing down. Were still moving forward with the B61-12 compatibility activities on different platforms.
In partnership with the National Nuclear Security Administration, Los Alamos National Laboratory and the Air Force, Sandia completed a B61-12 full-weapon system demonstration with the F-15E Strike Eagle fighter jet in March, and another in July with the Air Forces B-2 Spirit bomber.
Sandia is the design and engineering lab for non-nuclear components of the nations nuclear stockpile, including the B61-12. In addition to non-nuclear component development, Sandia serves as the technical integrator for the complete weapon, assuring the system meets requirements as a full-weapon system.
Showing the bombs real-world capability
During the Aug. 25 flight test, an F-35A flying faster than the speed of sound dropped a B61-12 containing non-nuclear and mock nuclear components from about 10,500 feet above Tonopah Test Range. The inert B61-12 struck the desert floor in the designated target area about 42 seconds later.
We successfully executed this historic, first-ever F-35A flight test at Tonopah Test Range within the specified delivery criteria, said Brian Adkins, range manager at TTR.
The success of this test, as with all other weapons evaluations, is only possible through the detailed planning, combined with full collaboration between TTR and the program engineers, and the execution of the test evolution by the field operators and recovery specialists in the combined team of Sandia and TTRs operations and maintenance subcontractor, Navarro Research and Engineering, he said.With the multiple phases and operational activities a test involves, the team at TTR is diligent to integrate safety and security into all segments to ensure proper precautions are implemented for mission success.
Coordination between Sandia, Los Alamos, the NNSA and the Air Force made the flight test possible, and initial data shows that all systems and interfaces between the refurbished bomb and the F-35A worked as expected.
Unlike previous fighter jets, the F-35A carries the bomb internally. The recent flight test was the first demonstration of a fully instrumented B61-12 release from an internal bomb bay on a fighter and the first such release at speeds of Mach 1 or greater, Samuels said.
This was the first test to exercise all systems, including mechanical, electrical, communication and release between the B61-12 and the F-35A, he said.
The test also came amidst now commonplace COVID-19 workplace restrictions, which can make planning more difficult but are not slowing down Sandias important mission work, said B61-12 program senior manager Christine Mitchell. Sandia National Labs, Los Alamos National Laboratory, NNSA and our Air Force partners are working diligently to ensure F-35A major milestones stay on track, despite the challenges presented by COVID-19.
The F-35A is a fifth generation fighter and is described by an Air Force online fact sheet as an agile, versatile, high-performance, 9-G capable multirole fighter with stealth technology and advanced sensors. Nine countries the United States, the United Kingdom, Italy, Netherlands, Turkey, Canada, Denmark, Norway and Australia were involved in the fighter jets development.
The latest test is a critical piece in the F-35A and B61-12 program, Samuels said. Aboard the newest fighter, the B61-12 provides a strong piece of the overall nuclear deterrence strategy for our country and our allies.
Sandia design and engineering is integral to B61-12 Life Extension Program
The compatibility testing is an essential part of the B61-12 Life Extension Program to refurbish, reuse or replace components, extend the bombs service life, and improve its safety, security and effectiveness.
A life extension program allows scientists and engineers to address the aging of nuclear weapons components. Some components are requalified and go back into a weapon without change; others that have aged are remanufactured using the original specifications; and sometimes the original technology is no longer available, so Sandia redesigns those parts using modern technology.
The first B61 entered service 50 years ago, and over the decades numerous modifications have been made to increase safety and reliability. The B61-12 consolidates and replaces most of the previous variants. The National Nuclear Security Administration recently announced plans to manufacture the first refurbished B61-12 in fiscal year 2022.
More here:
Flight tests to show B61-12 will work on Air Forces newest fighter jet - Newswise
Recommendation and review posted by Bethany Smith
Global Bilberry Extract Products Market To Witness Huge Gains Over 2020-2026 – The Courier
The globalBilberry Extract Productsmarket research report offers all the vital data in the domain. The latest report assists new bees as well as established market participants to analyze and predict the Bilberry Extract Products market at the regional as well as global level. It covers the volume [k MT] as well as revenues [USD Million] of the global Bilberry Extract Products market for the estimated period. Numerous key players GNC, Natures Way Products, LLC., Life Extension, Nutraceutical, Spring Valley, Source Naturals, Inc., Bluebonnet Nutrition, Biofinest, Swanson, Kanbo Natural Health Food, Nutrilite, Ahana Nutrition, Puritans Pride, Now foods, Natrol Vitamins & Supplements are dominating the global Bilberry Extract Products market. These players hold the majority of share of the global Bilberry Extract Products market.
Sample of global Bilberry Extract Products Market Report at::https://www.marketresearchstore.com/report/global-bilberry-extract-products-industry-market-report-2019-646924#RequestSample
The data presented in the global Bilberry Extract Products market offers budding opportunities, which help users to make strategic moves and prosper their business. The report highlights the impact of numerous factors that might result in obstructing or propelling the Bilberry Extract Products market at global as well as local level. The global Bilberry Extract Products market research report offers the summary of key players dominating the market including several aspects such as their financial summary, business strategy, and most recent developments in these firms.
Key points of the global Bilberry Extract Products market
Theoretical analysis of the global Bilberry Extract Products market stimulators, products, and other vital facets Recent, historical, and future trends in terms of revenue and market dynamics are reported Pin-point analysis of the competitive market dynamics and investment structure is predicted to grow Future market trends, latest innovations, and various business strategies are reported Market dynamics include growth influencers, opportunities, threats, challenges, and other crucial facets
The global Bilberry Extract Products market research report offers users with an all-inclusive package of market analysis that includes current market size, expansion rate, and value chain analysis. The global Bilberry Extract Products market is segmented on a regional basis Europe, North America, Latin America, Asia Pacific, and Middle East & Africa as well. To offer a comprehensive view and competitive outlook of the global Bilberry Extract Products market, our review team employs numerous methodological procedures, for instance, Porters five forces analysis.
Ask Any Kind of Query Before Purchasing the Report Here::https://www.marketresearchstore.com/report/global-bilberry-extract-products-industry-market-report-2019-646924#InquiryForBuying
This research report includes the analysis of various Bilberry Extract Products market segments {Tablets, Cheweables, Others}; {E-commerce, Medical Institutions, Supermarket}. The bifurcation of the global market is done based on its present and prospective inclinations. The regional bifurcation involves the present market scenario in the region along with the future projection of the global Bilberry Extract Products market. The global market report offers an overview of expected market conditions due to changes in the technological, topographical, and economic elements.
Questions answered in the report include
1. What is the expected market size by the end of the forecast period?2. What are the major factors initiating the global Bilberry Extract Products market growth?3. What are the latest developments and trending market strategies that are influencing the growth of the Bilberry Extract Products market?4. What are the key outcomes of the Bilberry Extract Products market developments?5. Who are the key players in the market?6. What are the opportunities and challenges faced by the key players?
Read Detailed Index of full Research Study at:https://www.marketresearchstore.com/report/global-bilberry-extract-products-industry-market-report-2019-646924
Key Reasons for Purchasing Global Bilberry Extract Products Market Report
New approaches and latest development trend that describe the structure of the market Advanced market breakdown structure Historical data and future market scope In-depth market analysis based on statistics, growth stimulators, and market developments Statistical data representation through figurative, numerical, and theoretical elaboration Report provides insight of the business and sales activities
Visit link:
Global Bilberry Extract Products Market To Witness Huge Gains Over 2020-2026 - The Courier
Recommendation and review posted by Bethany Smith
Traditional launch services may not suit the needs of the future Space Force – SpaceNews
The Space Forces launch enterprise is trying to gain better insight into the next wave of space innovation and figure out how the military could acquire those capabilities.
WASHINGTON SpaceX and United Launch Alliance were selected as U.S. national security launch providers based on their ability to deliver spacecraft to specific Earth orbits. How the Pentagon buys launch services in the future could change, however, as the military considers using emerging technologies and services known as space mobility and logistics.
Col. Robert Bongiovi, the director of the Space Forces launch enterprise, said his office is trying to gain better insight into the next wave of space innovation and figure out how the military could acquire those capabilities.
The Space and Missile Systems Center Launch Enterprise issued a request for information Nov. 10 asking companies to submit by Jan. 15 details on planned investments that would support space mobility and logistics.
Space tugs that move satellites to different orbits or within orbits, satellite refueling and servicing vehicles, and in-space manufacturing are some of the capabilities mentioned in the request for information as examples of the future space ecosystem.
These are all new space missions and capabilities that the military doesnt currently do. Bongiovi said last week at a Mitchell Institute event that the information submitted by the industry will help the Space Force decide on future investments in space access, mobility and logistics.
The Space Force in its vision document mentions space mobility and logistics as core competencies of the service.
Bongiovi said there are currently no plans to change the structure of the national security launch program, which relies on two launch providers to fly military and intelligence community satellites to multiple orbits. But he said the Space Force is doing market research that could inform the requirements for launch providers for the next national security space launch competition in 2024. Industry has a view of the future that is very expansive, he said.
We have to have honest conversations with industry on where theyre going and why, said Bongiovi. We also have to talk to our satellite providers and understand the demand.
Point-to-point transportation
One potential future use of space vehicles contemplated by the military is for cargo delivery on Earth. The U.S. Transportation Command, which is responsible for moving personnel and cargo around the world, is studying how space launch vehicles might be used to transport supplies and people in emergencies .
The idea has fascinated senior officials like Will Roper, assistant secretary for acquisitions for the Department of the Air Force. Using suborbital space vehicles for point-to-point delivery opens up intriguing possibilities, he told reporters Nov. 24.
My personal view, from what I see industry developing, is that logistics is a really interesting capability to rethink, he said. You can put mass quickly to the other side of the world. Special operations forces could be on the ground halfway around the world in minutes, said Roper.
Bongiovi seemed skeptical, however.
The Space Force launch enterprise, he said, supports studies on the use of space vehicles for cargo delivery. On whether this could actually be done, Bongiovi said, I dont think in the launch enterprise we have insight to answer this.
Launch companies will provide us the opportunity to take advantage of new uses for their systems, he said. I dont know that this is going to be one of them.
Lt. Gen. John Shaw, deputy commander of U.S. Space Command, suggested the military will wait and see how these technologies are employed in the private sector before it commits to fund programs.
Asked to explain how the military sees space mobility and logistics, Shaw said: Ive been intrigued by that concept for a long time.
I think we will watch it closely to see how effective it becomes on the commercial side, and in parallel see if we can introduce that into our architecture as a possible resilience measure or life extension measure, Shaw said Nov. 20 at an online event hosted by the Air Force Associations Schriever Chapter.
Shaw said the Space Force has to carefully analyze these options and figure out what parts of the space architecture benefit from that service.
More:
Traditional launch services may not suit the needs of the future Space Force - SpaceNews
Recommendation and review posted by Bethany Smith
Growing Value of Stem Cells in Medicine to Create a US$2,4 Billion Opportunity for Induced Pluripotent Stem Cell ((iPSC) – GlobeNewswire
New York, Nov. 25, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Global Induced Pluripotent Stem Cell (iPSC) Industry" - https://www.reportlinker.com/p05798831/?utm_source=GNW 4 billion by the year 2027, trailing a post COVID-19 CAGR of 6.6%, over the analysis period 2020 through 2027. Stem cells are undifferentiated cells that hold the capability to divide, and differentiate into specialized cells in the body. Stem cells act as repair system and replenish adult tissues, maintaining the turnover of regenerative organs such as the blood and skin. In organs, such as the bone marrow, stem cells frequently form replacement cells to repair the worn out tissue. These cells can respond to signals from the body and transverse a particular developmental pathway to differentiate into one specific cell type. Due to their regenerative properties, stem cells are being researched for therapeutic applications in diabetes, cardiovascular disease, neurodegenerative disease, cancer, autoimmune diseases, spinal cord defects, among others. Stem Cell research is an exciting field where continuous discoveries are being made on new sources of stem cells and new methods of their acquisition and harvesting. Of late, adult stem cells have garnered a lions share of the stem cell space, purely based on the fact that they require less expensive clinical trials, need to comply with fewer regulatory norms and ethical issues compared to other stem cell variants such as embryonic stem cells.
Researchers around the world have been focusing research activities to develop adult stem cell therapies in order to combat a variety of diseases ranging from diabetes to heart disease. Factually, adult stem cells are the only stem cells that have been approved for use in transplants for the treatment of diseases such as cancer. Interestingly, with drug development based on embryonic stem cells being challenged amid growing debate over ethics and regulation of this research, iPSCS offers an alternate step forward in the commercialization of stem cell therapies and regenerative medicine. Embryonic stem cell research continues to remain embroiled in ethical, religious, and political controversies across various countries around the world. Induced Pluripotent Stem Cells (iPSs), which are reprogrammed to mimic embryonic stem cell-like state allowing expression of genes and human cells needed for therapeutic purposes, offers an attractive alternate way forwarding in furthering the goals of stem cell research. Pioneered in 2006 and developed in the following year, these cells are created by conversion of somatic cells into PSCs by introducing certain genes including Myc, Klf4, Oct3/4 and Sox2.
Pluripotent stem cells hold tremendous potential in the regenerative medicine arena. Based on their ability to proliferate indefinitely and develop into desirable cell type such as heart, liver, neuronal and pancreatic cells, iPSCs offer a source of new cells that can replace lost or damaged cells. For instance, iPSCs can be developed into beta islet cells, blood cells or neuronal cells for the treatment of diabetes, leukemia and neurological disorders, respectively. Parkinsons, Alzheimers & spinal cord injuries are key neurologic diseases expected to benefit from iPS research. Dramatic rise in cancer cases worldwide and the need for novel anti-cancer therapies will emerge as a key driver for the growth of iPSCs. Interest in cancer research soars high on new hopes of direct reprogramming of cancer cells with enforced expression of pluripotency factors and the resulting dedifferentiation of transformed cancer cells. The ongoing pandemic is also opening up new opportunities for Human induced pluripotent stem cells (hiPSCs) by offering a reliable model for researchers involved in studying how coronavirus indirectly or directly affects different cells in the human body. Made from a small sample of blood or skin cells, hiPSCs are robust stem cells that can be developed into any cell type and then infected with the coronavirus in order to analyse the disease prognosis and the resulting effects. By deploying hiPSCs, researchers have identified that stem cell-derived cardiomyocytes (heart muscle cells) and blood vessels remain directly exposed to COVID-19 infection. Scientists identified that a significant portion of stem cell-derived cardiomyocytes ceased beating and expired within 3 days after being infected by coronavirus. Researchers can leverage the infected cardiomyocytes to screen for potential drug candidates that can restore their function and improve their survival; and also for identifying new antiviral drugs that potentially curtail coronavirus replication in the heart, reduce cardiac injury and curb the disease prognosis. Researchers can also utilize the infected cardiomyocytes to analyze COVID-induced myocarditis through addition of immune cells to their lab experiments.
Competitors identified in this market include, among others,
Read the full report: https://www.reportlinker.com/p05798831/?utm_source=GNW
I. INTRODUCTION, METHODOLOGY & REPORT SCOPE I-1
II. EXECUTIVE SUMMARY II-1
1. MARKET OVERVIEW II-1 Impact of Covid-19 and a Looming Global Recession II-1 Induced Pluripotent Stem Cells (iPSCs) Market Gains from Increasing Use in Research for COVID-19 II-1 Studies Employing iPSCs in COVID-19 Research II-2 Stem Cells, Application Areas, and the Different Types: A Prelude II-3 Applications of Stem Cells II-4 Types of Stem Cells II-4 Induced Pluripotent Stem Cell (iPSC): An Introduction II-5 Production of iPSCs II-6 First & Second Generation Mouse iPSCs II-6 Human iPSCs II-7 Key Properties of iPSCs II-7 Transcription Factors Involved in Generation of iPSCs II-7 Noteworthy Research & Application Areas for iPSCs II-8 Induced Pluripotent Stem Cell ((iPSC) Market: Growth Prospects and Outlook II-9 Drug Development Application to Witness Considerable Growth II-11 Technical Breakthroughs, Advances & Clinical Trials to Spur Growth of iPSC Market II-11 North America Dominates Global iPSC Market II-12 Competition II-12 Recent Market Activity II-13 Select Innovation/Advancement II-16
2. FOCUS ON SELECT PLAYERS II-17 Axol Bioscience Ltd. (UK) II-17 Cynata Therapeutics Limited (Australia) II-17 Evotec SE (Germany) II-17 Fate Therapeutics, Inc. (USA) II-17 FUJIFILM Cellular Dynamics, Inc. (USA) II-18 Ncardia (Belgium) II-18 Pluricell Biotech (Brazil) II-18 REPROCELL USA, Inc. (USA) II-18 Sumitomo Dainippon Pharma Co., Ltd. (Japan) II-19 Takara Bio, Inc. (Japan) II-19 Thermo Fisher Scientific, Inc. (USA) II-20 ViaCyte, Inc. (USA) II-20
3. MARKET TRENDS & DRIVERS II-21 Effective Research Programs Hold Key in Roll Out of Advanced iPSC Treatments II-21 Induced Pluripotent Stem Cells: A Giant Leap in the Therapeutic Applications II-21 Research Trends in Induced Pluripotent Stem Cell Space II-22 Exhibit 1: Worldwide Publication of hESC and hiPSC Research Papers for the Period 2008-2010, 2011-2013 and 2014-2016 II-22 Exhibit 2: Number of Original Research Papers on hESC and iPSC Published Worldwide (2014-2016) II-23 Concerns Related to Embryonic Stem Cells Shift the Focus onto iPSCs II-23 Regenerative Medicine: A Promising Application of iPSCs II-24 Induced Pluripotent: A Potential Competitor to hESCs? II-25 Exhibit 3: Global Regenerative Medicine Market Size in US$ Billion for 2019, 2021, 2023 and 2025 II-27 Exhibit 4: Global Stem Cell & Regenerative Medicine Market by Product (in %) for the Year 2019 II-27 Exhibit 5: Global Regenerative Medicines Market by Category: Breakdown (in %) for Biomaterials, Stem Cell Therapies and Tissue Engineering for 2019 II-28 Pluripotent Stem Cells Hold Significance for Cardiovascular Regenerative Medicine II-28 Exhibit 6: Leading Causes of Mortality Worldwide: Number of Deaths in Millions & % Share of Deaths by Cause for 2017 II-30 Leading Causes of Mortality for Low-Income and High-Income Countries II-30 Growing Importance of iPSCs in Personalized Drug Discovery II-31 Persistent Advancements in Genetics Space and Subsequent Growth in Precision Medicine Augur Well for iPSCs Market II-33 Exhibit 7: Global Precision Medicine Market (In US$ Billion) for the Years 2018, 2021 & 2024 II-34 Increasing Prevalence of Chronic Disorders Supports Growth of iPSCs Market II-34 Exhibit 8: Worldwide Cancer Incidence: Number of New Cancer Cases Diagnosed for 2012, 2018 & 2040 II-35 Exhibit 9: Number of New Cancer Cases Reported (in Thousands) by Cancer Type: 2018 II-36 Exhibit 10: Fatalities by Heart Conditions: Estimated Percentage Breakdown for Cardiovascular Disease, Ischemic Heart Disease, Stroke, and Others II-37 Exhibit 11: Rising Diabetes Prevalence Presents Opportunity for iPSCs Market: Number of Adults (20-79) with Diabetes (in Millions) by Region for 2017 and 2045 II-38 Aging Demographics Add to the Global Burden of Chronic Diseases, Presenting Opportunities for iPSCs Market II-38 Exhibit 12: Expanding Elderly Population Worldwide: Breakdown of Number of People Aged 65+ Years in Million by Geographic Region for the Years 2019 and 2030 II-39 Growth in Number of Genomics Projects Propels Market Growth II-39 Genomic Initiatives in Select Countries II-40 Exhibit 13: New Gene-Editing Tools Spur Interest and Investments in Genetics, Driving Lucrative Growth Opportunities for iPSCs: Total VC Funding (In US$ Million) in Genetics for the Years 2014, 2015, 2016, 2017 and 2018 II-41 Launch of Numerous iPSCs-Related Clinical Trials Set to Benefit Market Growth II-41 Exhibit 14: Number of Induced Pluripotent Stem Cells based Studies by Select Condition: As on Oct 31, 2020 II-43 iPSCs-based Clinical Trial for Heart Diseases II-43 Induced Pluripotent Stem Cells for Stroke Treatment II-44 ?Off-the-shelf? Stem Cell Treatment for Cancer Enters Clinical Trial II-44 iPSCs for Hematological Disorders II-44 Market Benefits from Growing Funding for iPSCs-Related R&D Initiatives II-44 Exhibit 15: Stem Cell Research Funding in the US (in US$ Million) for the Years 2016 through 2021 II-46 Human iPSC Banks: A Review of Emerging Opportunities and Drawbacks II-46 Human iPSC Banks Worldwide: An Overview II-48 Cell Sources and Reprogramming Methods Used by Select iPSC Banks II-49 Innovations, Research Studies & Advancements in iPSCs II-50 Key iPSC Research Breakthroughs for Regenerative Medicine II-50 Researchers Develop Novel Oncogene-Free and Virus-Free iPSC Production Method II-51 Scientists Study Concerns of Genetic Mutations in iPSCs II-52 iPSCs Hold Tremendous Potential in Transforming Research Efforts II-52 Researchers Highlight Potential Use of iPSCs for Developing Novel Cancer Vaccines II-54 Scientists Use Machine Learning to Improve Reliability of iPSC Self-Organization II-54 STEMCELL Technologies Unveils mTeSR? Plus II-55 Challenges and Risks Related to Pluripotent Stem Cells II-56 A Glance at Issues Related to Reprogramming of Adult Cells to iPSCs II-57 A Note on Legal, Social and Ethical Considerations with iPSCs II-58
4. GLOBAL MARKET PERSPECTIVE II-59 Table 1: World Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-59
Table 2: World 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets for Years 2020 & 2027 II-60
Table 3: World Current & Future Analysis for Vascular Cells by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-61
Table 4: World 7-Year Perspective for Vascular Cells by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-62
Table 5: World Current & Future Analysis for Cardiac Cells by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-63
Table 6: World 7-Year Perspective for Cardiac Cells by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-64
Table 7: World Current & Future Analysis for Neuronal Cells by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-65
Table 8: World 7-Year Perspective for Neuronal Cells by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-66
Table 9: World Current & Future Analysis for Liver Cells by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-67
Table 10: World 7-Year Perspective for Liver Cells by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-68
Table 11: World Current & Future Analysis for Immune Cells by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-69
Table 12: World 7-Year Perspective for Immune Cells by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-70
Table 13: World Current & Future Analysis for Other Cell Types by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-71
Table 14: World 7-Year Perspective for Other Cell Types by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-72
Table 15: World Current & Future Analysis for Cellular Reprogramming by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-73
Table 16: World 7-Year Perspective for Cellular Reprogramming by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-74
Table 17: World Current & Future Analysis for Cell Culture by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-75
Table 18: World 7-Year Perspective for Cell Culture by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-76
Table 19: World Current & Future Analysis for Cell Differentiation by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-77
Table 20: World 7-Year Perspective for Cell Differentiation by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-78
Table 21: World Current & Future Analysis for Cell Analysis by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-79
Table 22: World 7-Year Perspective for Cell Analysis by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-80
Table 23: World Current & Future Analysis for Cellular Engineering by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-81
Table 24: World 7-Year Perspective for Cellular Engineering by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-82
Table 25: World Current & Future Analysis for Other Research Methods by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-83
Table 26: World 7-Year Perspective for Other Research Methods by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-84
Table 27: World Current & Future Analysis for Drug Development & Toxicology Testing by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-85
Table 28: World 7-Year Perspective for Drug Development & Toxicology Testing by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-86
Table 29: World Current & Future Analysis for Academic Research by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-87
Table 30: World 7-Year Perspective for Academic Research by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-88
Table 31: World Current & Future Analysis for Regenerative Medicine by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-89
Table 32: World 7-Year Perspective for Regenerative Medicine by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-90
Table 33: World Current & Future Analysis for Other Applications by Geographic Region - USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 II-91
Table 34: World 7-Year Perspective for Other Applications by Geographic Region - Percentage Breakdown of Value Sales for USA, Canada, Japan, China, Europe, Asia-Pacific and Rest of World for Years 2020 & 2027 II-92
III. MARKET ANALYSIS III-1
GEOGRAPHIC MARKET ANALYSIS III-1
UNITED STATES III-1 Table 35: USA Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-1
Table 36: USA 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-2
Table 37: USA Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-3
Table 38: USA 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-4
Table 39: USA Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-5
Table 40: USA 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-6
CANADA III-7 Table 41: Canada Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-7
Table 42: Canada 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-8
Table 43: Canada Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-9
Table 44: Canada 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-10
Table 45: Canada Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-11
Table 46: Canada 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-12
JAPAN III-13 Table 47: Japan Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-13
Table 48: Japan 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-14
Table 49: Japan Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-15
Table 50: Japan 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-16
Table 51: Japan Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-17
Table 52: Japan 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-18
CHINA III-19 Table 53: China Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-19
Table 54: China 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-20
Table 55: China Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-21
Table 56: China 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-22
Table 57: China Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-23
Table 58: China 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-24
EUROPE III-25 Table 59: Europe Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Geographic Region - France, Germany, Italy, UK and Rest of Europe Markets - Independent Analysis of Annual Sales in US$ Thousand for Years 2020 through 2027 III-25
Table 60: Europe 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Geographic Region - Percentage Breakdown of Value Sales for France, Germany, Italy, UK and Rest of Europe Markets for Years 2020 & 2027 III-26
Table 61: Europe Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-27
Table 62: Europe 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-28
Table 63: Europe Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-29
Table 64: Europe 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-30
Table 65: Europe Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-31
Table 66: Europe 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-32
FRANCE III-33 Table 67: France Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-33
Table 68: France 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-34
Table 69: France Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-35
Table 70: France 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Research Method - Percentage Breakdown of Value Sales for Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods for the Years 2020 & 2027 III-36
Table 71: France Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Application - Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-37
Table 72: France 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Application - Percentage Breakdown of Value Sales for Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine and Other Applications for the Years 2020 & 2027 III-38
GERMANY III-39 Table 73: Germany Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-39
Table 74: Germany 7-Year Perspective for Induced Pluripotent Stem Cell (iPSC) by Cell Type - Percentage Breakdown of Value Sales for Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells and Other Cell Types for the Years 2020 & 2027 III-40
Table 75: Germany Current & Future Analysis for Induced Pluripotent Stem Cell (iPSC) by Research Method - Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering and Other Research Methods - Independent Analysis of Annual Sales in US$ Thousand for the Years 2020 through 2027 III-41
Please contact our Customer Support Center to get the complete Table of ContentsRead the full report: https://www.reportlinker.com/p05798831/?utm_source=GNW
About ReportlinkerReportLinker is an award-winning market research solution. Reportlinker finds and organizes the latest industry data so you get all the market research you need - instantly, in one place.
__________________________
See the rest here:
Growing Value of Stem Cells in Medicine to Create a US$2,4 Billion Opportunity for Induced Pluripotent Stem Cell ((iPSC) - GlobeNewswire
Recommendation and review posted by Bethany Smith
The stem/progenitor landscape is reshaped in a mouse model of essential thrombocythemia and causes excess megakaryocyte production – Science Advances
INTRODUCTION
The myeloproliferative neoplasms are a family of clonal blood disorders characterized by overproduction of platelets [essential thrombocythemia (ET)], overproduction of red blood cells [polycythemia vera (PV)], or bone marrow fibrosis [myelofibrosis (MF)]. The genetic bases for these diseases have largely been described: Mutations in JAK2 are found in 99% of PV and 50 to 60% of ET and MF cases, while frameshift mutations in CALR are responsible for 25 to 40% of cases of ET and MF (13). Frameshift mutants of calreticulin (CALR) have a novel C terminus that acts as a rogue ligand for the thrombopoietin receptor, MPL, and activates Janus kinasesignal transducer and activator of transcription (JAK-STAT) signaling (4, 5). We recently described the generation of a mouse model of mutant CALR-driven ET that faithfully recapitulates the key phenotypes of the human disease, namely, increased numbers of cells throughout the megakaryocytic (MK) lineage, particularly platelets (6).
Hematopoiesis is classically modeled as a stepwise process beginning with a multipotent hematopoietic stem cell (HSC), which is functionally defined by its capability to reconstitute multilineage hematopoiesis when transplanted into a myeloablated recipient (7). This HSC then transits through a series of intermediate stages with increasing lineage restriction to terminally differentiated blood cells (8, 9). However, newly popularized single-cell technologies such as single-cell RNA sequencing (scRNAseq) have reshaped our understanding of hematopoiesis and suggest that cells travel through a continuum of differentiation rather than a series of rigidly defined stages (10, 11). In a recent demonstration of the power of scRNAseq to untangle complex differentiation processes, it was used to interrogate the transcriptomes of hematopoietic stem and progenitor cells (HSPCs) to identify novel intermediate populations within erythropoiesis, which could then be isolated and characterized via fluorescence-activated cell sorting (FACS) strategies (12).
While HSCs are traditionally defined to be capable of reconstituting all blood lineages in transplantation experiments, there is an increasing body of evidence that some cells within the immunophenotypic HSC compartment already exhibit some lineage bias or restriction (1315). Studies in mice have shown that MK and erythroid lineages may branch off before other myeloid and lymphoid lineages (1618), and lineage tracing studies have shown the MK lineage to be the earliest generated from HSCs (1923). A transposon-based lineage tracing strategy showed some tags to be shared between long-term HSCs (LT-HSCs) and megakaryocyte progenitors (MkPs) but not multipotent progenitors (MPPs), indicative of a direct pathway linking HSCs and MK bypassing MPP (19). We therefore asked whether our mouse model of mutant CALR-driven ET could allow us to interrogate the differences in the hematopoietic landscapes between wild-type (WT) and disease model mice, with a particular focus on MK trajectories.
We generated scRNAseq data from FACS-sorted HSPCs [Lin Sca1+ cKit+ (LSK) and Lin Sca1 cKit+ (LK) populations] from a pair of WT and CALR DEL (knock-in of del52 allele) homozygous (HOM) littermate mice. After quality control, we retained 11,098 WT (5959 LSK and 5139 LK) and 15,547 HOM (7732 LSK and 7815 LK) cells for downstream analysis. We began by defining highly variable genes, which we used to perform principal component analysis (PCA) and generate a k = 7 nearest-neighbor graph. Cells were then assigned to clusters by mapping onto a previously published dataset of 44,082 LK cells (24), with manual annotation of clusters (fig. S1A). Cells from all major blood lineages can be seen and separate into distinct trajectories. To determine which cells were over- or underrepresented in the CALR DEL HOM mouse, we compared relative numbers of cells from each genotype. The most notable changes in relative cell abundance were increased numbers of cells in the HSC and MK clusters (fig. S1B), consistent with the increased platelet phenotype of our ET mouse model (6). We repeated the analysis on a second pair of WT and CALR DEL HOM littermate mice, in this case retaining 3451 WT (972 LSK and 2479 LK) and 12,372 HOM (4548 LSK and 7824 LK) cells for downstream analysis after quality control, and again observed an increase in cells in the HSC and MK clusters (fig. S1C).
To better understand the subgroups of cells within stem/progenitor cells, we chose to use partition-based graph abstraction (PAGA) (25) to visualize our data. This method generates a graph in which each node represents a group of closely related cells and edge weights correspond to the strength of connection between two nodes. We again compared relative abundances between WT and CALR DEL HOM mice and colored the nodes so red nodes are enriched in CALR mice, while blue nodes are underrepresented. We observed that the fine cluster that was most overrepresented in CALR DEL HOM mice (marked with an arrow) fell between the HSC and MK clusters in both repeats (Fig. 1A and fig. S1D). We plotted the expression of the MK markers Cd9, Itga2b (CD41), Mpl, Pf4, and VWF in our PAGA and hypothesized two MK trajectories, as indicated by the green and blue arrows (fig. S1E). As the fine cluster most overrepresented in CALR DEL HOM mice would be an intermediate on one of these trajectories (green arrow), we further hypothesized that these cells would be of particular relevance in the disease setting of mutant CALR-driven ET and thus aimed to further study them.
(A) PAGA of scRNAseq data from WT and CALR DEL HOM mice. Red nodes represent those present at increased abundance in CALR DEL HOM mice, while blue nodes represent those at reduced abundance. The most highly enriched node is noted with an arrow. (B) RNA expression of the flow cytometry markers CD48, EPCR (Procr), and CD150 (Slamf1) plotted on PAGA graphs from (A). Cells within our node of interest (marked with an arrow) are CD48, EPCR, and CD150+. (C) Representative plots of SLAM cells from WT and CALR DEL HOM mice. CALR DEL HOM mice show higher numbers of both ESLAMs (Lin CD48 CD150+ CD45+ EPCR+) and pMKPs (Lin CD48 CD150+ CD45+ EPCR). FITC, fluorescein isothiocyanate; PE, phycoerythrin. (D) Quantification of bone marrow frequency of pMKPs in WT and CALR DEL HOM mice. The frequency of pMKPs within live bone marrow mononuclear cells (BMMNCs) is significantly increased in CALR DEL HOM mice (WT, n = 3, 0.00029 0.00008; HOM, n = 3, 0.0025 0.0008; *P = 0.042).
We examined the expression of a series of genes typically used to FACS isolate different hematopoietic populations and found this fine cluster to be CD48, EPCR (Procr), and CD150+ (Slamf1) (Fig. 1B). We designed an immunophenotypic scheme to identify and isolate cells from this fine cluster, defining them to be Lin, CD150+, CD48, EPCR, and CD45+. On the basis of our subsequent characterization of these cells, we eventually termed them proliferative MkPs or pMKPs. Consistent with our transcriptomic data, when comparing WT mice to CALR mutant mice, we found an increase in the frequency of pMKPs in CALR DEL HOM mice as assayed by flow cytometry (Fig. 1, C and D). We also found that pMKPs were expanded in CALR DEL HET mice, albeit to a lesser extent than observed in CALR DEL HOM mice (fig. S1F).
To characterize pMKPs, we FACS-sorted single ESLAM (EPCR+ SLAM) HSCs (Lin CD45+ CD48 CD150+ EPCR+) (26), pMKPs (Lin CD45+ CD48 CD150+ EPCR), and MkPs (Lin Sca1 cKit+ CD41+ CD150+) (27) (fig. S2A) from WT mice into individual wells of a 96-well plate and observed them every day for 4 days. We analyzed our sort data and observed that in pMKPs, markers traditionally used to define MkPs were Sca1/lo/mid, cKit+, and CD41mid/+ (fig. S2B). pMKPs were additionally CD9+ and MPL+ (fig. S2C). On each day, we classified each well with surviving cell(s) into one of four categories, using cell size as a proxy for megakaryopoiesis (2830): (i) exactly one large cell, presumed to be a megakaryocyte; (ii) multiple large cells; (iii) mixed expansion, with both large and small cells; and (iv) expansion with only small cells (Fig. 2A). To verify that larger cells represented MK cells, using cells from day 4 ESLAM, pMKP, and MkP colonies, we quantified average CD41 intensity via immunofluorescence and classified cells as small or large via bright-field microscopy, using a small/large dichotomy assessed via bright-field microscopy to match the classification scheme used in Fig. 2A. Here, we confirmed that large cells have significantly higher CD41 staining, supporting their identification as MK (fig. S2D). In some cases, particularly large cells within mixed colonies showed very high CD41 staining and membrane extensions that resembled proplatelets (representative picture is shown in fig. S2E). Furthermore, we sorted pMKPs from VWF (von Willebrand factor)green fluorescent proteinpositive (GFP+) mice and found that large cells had a very bright VWF-GFP signal, supporting their identification as MK. Smaller cells in these clones had a much dimmer VWF-GFP signal, suggesting that they likely represent more immature cells that have not progressed as far through megakaryopoiesis (fig. S2F).
(A) Representative pictures of in vitro culture output of single ESLAMs, pMKPs, and MkPs into four categories: 1 MK, >1 MK, mixed, or proliferation only. (B) Classification of in vitro culture output of single ESLAMs, pMKPs, and MkPs at day 4 after FACS isolation. ESLAMs almost exclusively proliferated without producing megakaryocytes, while MkPs almost exclusively produced MKs, usually producing only a single MK. pMKPs showed a strong MK bias but were more likely to proliferate than were MkPs. ESLAMs, n = 306 wells from five experiments; pMKPs, n = 291 wells from six experiments; MkPs, n = 235 wells from five experiments. Chi-square test, ****P < 0.0001. (C) Timing of megakaryopoiesis in ESLAMs, pMKPs, and MkPs. Individual cells were observed for 4 days after sort, and the first date on which cell(s) showed signs of megakaryopoiesis was noted. MkPs were faster to begin megakaryopoiesis than were pMKPs (at day 2, MkPs: 89.5 0.7%; pMKPs: 50 6%; *P = 0.02). ESLAMs, n = 5; pMKPs, n = 6; MkPs, n = 5. (D) Log2-transformed cell counts of megakaryocytes from pMKPs and MkPs after 4 days of culture. Each point represents the average value from one of four separate experiments. Average of four experiments: pMKP, 1.12; MkP, 0.412, *P = 0.0295. (E) Histogram of the minimum number of cell divisions for 103 pMKPs and 158 MkPs that produced only megakaryocytes after 4 days of culture across four experiments. Chi-square test, ***P = 0.0001.
The vast majority of ESLAMs showed expansion with only small cells at day 4, consistent with being highly primitive HSCs with considerable proliferative potential, but not yet producing megakaryocytes. Similarly, as predicted for MkPs, more than 95% of wells showed exclusively production of MKs at day 4, with the majority producing only one MK. This lack of in vitro proliferation for single MkPs is consistent with previously published results, where 75% of MkPs did not divide and none produced more than 10 MKs (31). pMKPs exhibited an intermediate phenotype: While approximately 90% of wells showed production of some MKs, they were much more likely to produce multiple MK than were MkPs. In particular, pMKPs frequently proliferated into mixed colonies with both large and small cells, a behavior that was rarely seen for either ESLAMs or MkPs (Fig. 2B). Kinetic analysis showed that MkPs were faster to begin megakaryopoiesis than were pMKPs (Fig. 2C), and when considering only wells that produced only MKs, pMKPs produced more MKs than did MkPs (Fig. 2, D and E). pMKPs maintained their MK bias even when incubated under pro-erythroid or pro-myeloid conditions (fig. S3A). Culturing cells with thrombopoietin (THPO) increased the proportion of pMKPs that formed colonies with multiple MKs while reducing the number of mixed colonies (fig. S3B). To verify that our observed MK bias is not simply due to culture conditions supporting only megakaryopoiesis, we cultured ESLAMs under the same conditions for 10 days followed by flow cytometric analysis and observed multilineage differentiation (fig. S3C).
To examine the extent of overlap between our pMKPs and traditionally defined MkPs, we stained bone marrow with a panel incorporating all necessary markers and index sorted single pMKPs and MkPs. On the basis of index sort values, 97% of MkPs were CD45+, 50% were EPCR, and only 2% were CD48; when taken together, fewer than 1% of immunophenotypic MkPs also fell within the pMKP gate (fig. S3D); thus, pMKPs and MkPs can be FACS-separated on the basis of CD48 and EPCR. In contrast, we found that an average of 51% of pMKPs were also immunophenotypically MkPs (CD41+ Sca1 cKit+) (fig. S3E). As we observed a partial overlap between pMKPs and MkPs, we used our index sort data to assign each pMKP an overlap score based on the levels of CD41, Sca1, and cKit: 1/3 if only one marker overlapped, 2/3 if two overlapped, and 3/3 for pMKPs that also fall within the MkP immunophenotypic gate. No pMKPs had an overlap score of 0/3. We used the same classification scheme as in Fig. 2B and found that lower overlap scores correlated to a more proliferative, less MK-restricted phenotype: The pMKPs that are least similar to MkPs are the most proliferative and the least restricted to the MK lineage, although they still display a strong preference for MK production (fig. S3F). pMKPs with the lowest overlap score took the longest to enter megakaryopoiesis (fig. S3G). Together, our data indicate that pMKPs represent a group of cells with an MK bias and an increased proliferative potential as compared to traditionally defined MkPs.
We next determined whether pMKPs were capable of producing platelets in vivo. We made use of CD45.2 VWF-GFP donor mice and cKit W41/W41 CD45.1 recipient mice, which allowed us to track platelets (via VWF-GFP) and nucleated cells (by CD45.1/CD45.2 staining) (Fig. 3A). We FACS-sorted ESLAMs, pMKPs, and MkPs from VWF-GFP donor mice and transplanted 30, 60, or 120 cells per recipient into sublethally irradiated W41 mice along with 250,000 spleen MNCs (mononuclear cells) (SPMNCs) as helper cells and assayed peripheral blood chimerism every week for 4 weeks and at 16 weeks. We did not sort on VWF-GFP+ at this stage, but flow cytometry analysis showed that ESLAMs, pMKPs, and MkPs were all highly enriched for VWF-GFP expression when compared to total bone marrow (fig. S4A). We also transplanted one mouse per cohort with 250,000 SPMNCs alone to serve as a negative control to help with gating to avoid false positives. Representative gating strategies are shown in fig. S4 (B and C). As expected, ESLAMs were able to generate relatively high levels of platelets at all three cell doses, starting with a very low level at week 1 and increasing over the course of 4 weeks and continuing up to 16 weeks (although one recipient of 30 ESLAMs was lost to follow-up before the 16-week time point). pMKPs and MkPs were only able to reconstitute platelets at a very low level (1/105 to 1/104), even at the highest cell dose (Fig. 3, B to D and summarized in E). Low levels of donor-derived platelets were detected in 10 of 12 pMKP recipients and 8 of 13 MkP recipients within the first 4 weeks; extended observation up to 16 weeks showed that few recipients continued to produce VWF-GFP+ platelets, although all 3 pMKP recipients at the highest dose still showed VWF-GFP+ platelets. ESLAMs successfully produced CD11b+ myeloid cells in 10 of 10 recipients across varying cell doses, while pMKPs and MkPs only produced CD11b+ cells at a low level in 3 of 12 and 2 of 10 recipients, respectively (fig. S4, D to F and summarized in G). Therefore, we concluded that while pMKPs and MkPs have limited capabilities in a transplantation experiment, they both show an MK bias, in agreement with their in vitro behaviors. These low levels of reconstitution suggest that pMKPs and MkPs do not divide considerably in vivo, again similar to in vitro data.
(A) Schematic of VWF-GFP+ transplantation strategy. ESLAMs, pMKPs, and MkPs were sorted from VWF-GFP+, CD45.2 donor mice and transplanted into sublethally irradiated cKit W41/W41 CD45.1 recipients. PB, peripheral blood. (B) Platelet reconstitution from 30 donor cells. (C) Platelet reconstitution from 60 donor cells. (D) Platelet reconstitution from 120 donor cells. (E) Table summarizing numbers of mice with successful platelet production from ESLAMs, pMKPs, and MkPs. Here, transplanted cells were defined to have produced platelets if platelets were observed at a level of at least 1 in 105 at one or more time points within the first 4 weeks after transplantation.
Our single-cell transcriptomic analysis showed pMKPs to be an intermediate stage on an MK trajectory maintaining CD48 negativity (Fig. 1B and green arrow in fig. S1E), which suggests that they bypass the traditional MPP2 pathway (blue arrow in fig. S1E). We therefore asked whether we could show production of pMKPs from HSCs in an MPP2-independent manner by making use of a mouse model allowing inducible depletion of HSPCs. In this model, Tal1-Cre/ERT mice are crossed with R26DTA mice, wherein treatment with tamoxifen leads to specific expression of diphtheria toxin in HSCs and primitive progenitors and hence suicidal depletion of these early populations (Fig. 4A) (32). Within 6 weeks after HSC depletion, very few LT-HSCs remain, but levels of MPPs, committed progenitors, and mature blood cells are only slightly lower than in control animals (32). We reasoned that if pMKPs arise directly from HSCs, they should be depleted to a similar extent as HSCs, while if they arise from an MPP pathway, they should be depleted to a similar extent as MPPs (i.e., to a lesser extent than HSCs).
(A) Schematic of DTA (diphtheria toxin fragment A) HSC depletion model experiment. Tal1-CreERT/R26DTA mice were treated with four doses of tamoxifen at 0.1 mg/g to induce suicidal depletion of HSCs and then euthanized after 6 weeks for bone marrow (BM) analysis. (B) Frequencies of stem and progenitor cells with or without stem cell depletion. Cell populations that were significantly diminished by suicidal depletion of HSCs include ESLAMs (Cre, 17.1 10.8/105 BMMNC; Cre+, 4.3 2.0/105 BMMNC; *P = 0.012), LTHSCs (LSK CD48 CD150+) (Cre, 15 12/105 BMMNC; Cre+, 3.6 1.7/105 BMMNC; *P = 0.031), pMKPs (Cre, 13.0 7.6/105 BMMNC; Cre+, 4.1/105 BMMNC; *P = 0.013), and MkPs (Cre, 44.2 26.4/105 BMMNC; Cre+, 21.4 6.1/105 BMMNC; *P = 0.046); Cre, n = 8 and Cre+, n = 10. MPP2 (Cre, 25.1 29.1/105 BMMNC; Cre+, 13.3 3.6/105 BMMNC; P = 0.48) and preMegE (Cre, 90.0 62.9/105 BMMNC; Cre+, 73.9 29.6/105 BMMNC; P = 0.66) populations were depleted to lesser extents that did not reach statistical significance; Cre n = 4 and Cre+ n = 6. ns, not significant.
We compared mice carrying either no Cre or Tal1-Cre/ERT after treatment with tamoxifen to induce specific depletion of HSCs. We observed a depletion of approximately 75% in the numbers of HSCs [whether using ESLAM markers or LT-HSC (LSK CD48 CD150+) markers] and a 68% reduction in the numbers of pMKPs in HSC-depleted mice. By contrast, there was no significant reduction in MPP2 or preMegE populations, while MkPs were reduced by approximately 51% (Fig. 4B). Consistent with previously published results, we observed no statistically significant reduction in other multipotent populations, including MPP3 and MPP4 (33), and committed progenitor populations, including CFU-E (erythroid colony-forming units), pCFU-E, pGM (pre-granulocyte/macrophage), and GMP (granulocyte/monocyte progenitors) (fig. S5) (27). We noted that one Cre mouse was an outlier, with noticeably higher frequencies of almost all progenitor populations, and tested removing this outlier to ensure our conclusions were not unduly relying on this mouse. With the outlier removed, we calculated reductions of 68% in ESLAMs (P = 0.0001), 60% in pMKPs (P = 1.5 105), and an increase of 24% in MPP2 (P = 0.50). Our analysis is therefore robust to the removal of this outlier and demonstrates that the reduction in pMKP levels correlates more closely to that of ESLAMs than that of MPP2. Together, these data support a model in which pMKPs are produced from HSCs in an MPP2-independent manner and MkPs can be generated from pMKPs or via MPP2, accounting for their intermediate level of reduction.
After characterizing the pMKP population in WT mice, we next asked whether there were qualitative differences between WT and CALR DEL HOM cells along the MK trajectory and not solely a quantitative difference. To do so, we sorted single ESLAMs, pMKPs, and MkPs from WT and CALR DEL HOM mice and monitored their in vitro behavior over 4 days. While very few WT ESLAMs showed any MKs within the first 4 days after sort, a higher proportion of CALR DEL HOM ESLAMs showed MKs within mixed colonies (Fig. 5A). CALR DEL HOM pMKPs showed similar proportions of wells in each category (Fig. 5B), while CALR DEL HOM MkPs were more likely to form multiple MKs and less likely to form a single MK (Fig. 5C). To assess the statistical significance of these differences, using a Fishers exact or chi-square test required consolidation of our data into fewer categories, as some categories contained values that were too low (for example, for day 4 ESLAMs, the categories 1 MK and >1 MK were 0 in both WT and HOM). We thus consolidated ESLAM data into two categoriesno MK and MK (Fig. 5D)and pMKP and MkP data into three categories1 MK, >1 MK, and mixed + prolif only (Fig. 5, E and F). This showed that CALR DEL HOM ESLAMs were significantly more likely to form MKs (Fig. 5D). CALR DEL HOM pMKPs showed no statistically significant difference, suggesting no change in their MK bias or proliferative behavior compared to WT pMKPs (Fig. 5E). CALR DEL HOM MkPs were significantly more proliferative than were WT MkPs (Fig. 5F). We also extended our observation of ESLAM clones to day 7 and observed an even stronger increase in the production of megakaryocytes from CALR DEL HOM ESLAMs, an increase noted both in wells producing mixed clones and in those producing MK-only clones (Fig. 5, G and H).
(A) Classification of in vitro culture output of single ESLAMs from WT and CALR DEL HOM mice at day 4, using the classification scheme as in Fig. 2A. WT, n = 223; HOM, n = 225. (B) Classification of in vitro culture output of single pMKPs from WT and CALR DEL HOM mice at day 4; WT, n = 117; HOM, n = 161. Chi-square test P = 0.9201. (C) Classification of in vitro culture output of single MkPs from WT and CALR DEL HOM mice at day 4; WT, n = 136; HOM, n = 152. (D) Reclassification of data from (A) into two categories (MK or no MK) for a Fishers exact test, *P = 0.0191. (E) Reclassification of data from (B) into three categories (1 MK, >1 MK, and mixed + prolif only) for a chi-square test, P = 0.8183. (F) Reclassification of data from (C) into three categories (1 MK, >1 MK, and mixed + prolif only) for a chi-square test, **P = 0.0069. (G) Classification of in vitro culture output of single ESLAMs at day 7; WT, n = 136; HOM, n = 152. (H) Reclassification of data from (G) into two categories (MK or no MK) for a Fishers exact test, **P = 0.0014. (I) pMKPs as a proportion of live cells generated from in vitro culture of WT and CALR DEL HOM ESLAMs, assessed at day 3. WT, 0.062 0.015; HOM, 0.193 0.036, *P = 0.0135, n = 3 independent mice.
We also considered log2-transformed cell counts from those wells with exclusively megakaryocytes (i.e., 1 MK and >1 MK). In some cases, we observed the death of a cell or cells over our 4-day observation period; to account for cell death, we used the maximum number of cells observed over these 4 days. Mann-Whitney U tests showed no significant difference for pMKPs but a significant increase in MK production from CALR DEL HOM MkPs (fig. S6, A and B). Similarly, calculations of the minimum number of divisions required to produce the observed number of MKs found no difference for pMKPs but a significant shift to more divisions from CALR DEL HOM MkPs (fig. S6, C and D). We also cultured ESLAMs in vitro and assayed for the production of pMKPs, finding that CALR DEL HOM ESLAMs gave rise to significantly more pMKPs than did their WT counterparts (Fig. 5I). Together, we conclude that CALR DEL is acting at multiple stages of megakaryopoiesis, promoting an MK bias from the earliest HSC compartments and increased proliferation at both HSC and MkP levels. While pMKPs are increased in number in CALR DEL HOM mice, these cells do not show altered proliferation or MK bias in vitro.
Last, we made use of our scRNAseq data to compare gene expression between WT and CALR DEL HOM cells along the MK trajectory. We considered cells within 2 of the 13 clusters defined by our transcriptomic data (HSC and MK; fig. S1A) and 1 fine cluster (pMKP; arrow in Fig. 1A) (Fig. 6, A to C). As the pMKP fine cluster had fewer cells (24 in WT and 247 in CALR DEL HOM) than the larger HSC and MK clusters, we were only able to confidently call a small number of differentially expressed genes (DEGs) within this cluster. We performed Ingenuity Pathway Analysis (IPA) to determine which biological pathways and upstream regulators were most affected in the HSC and MK clusters; the small numbers of DEGs in pMKPs resulted in no statistically significant hits via IPA. The most affected canonical pathways fell into three broad groups: cell cycle (in blue), unfolded protein response (gold), and cholesterol biosynthesis (green) (Fig. 6, D and E). Full lists of canonical pathways, P values, and z scores are available in tables S1 (HSC) and S2 (MK). Genes contributing to these three pathways are highlighted in the same colors in Fig. 6, A to C; we note that pMKPs also show up-regulation of several UPR (unfolded protein response)associated genessuch as Hspa5, Pdia3, and Pdia6in addition to two known STAT targets (Ifitm2 and Socs2).
(A to C) Volcano plots showing DEGs between WT and CALR DEL HOM cluster 3 (HSC) (A), pMKP fine cluster (B), and cluster 11 (MK) (C). Genes within certain representative Gene Ontology (GO) terms are colored: regulation of cholesterol biosynthetic process (GO:0045540) (green), response to ER stress (GO:0034976) (gold), and regulation of mitotic cell cycle (GO:0007346) (blue). Other DEGs are colored in red. (D and E) Bar graphs showing z scores for up-regulated canonical pathways in cluster 3 (HSC) (C) and cluster 11 (MK) (D), filtered by P < 0.01 and z score of >1 or <1. Bars are highlighted in green for cholesterol biosynthesis, gold for ER stress/unfolded protein response, or blue for cell cycle. (F) Upstream regulator analysis. Hits were filtered by P < 0.01. Bar graph showing the 10 most up-regulated and 10 most down-regulated predicted upstream regulators, when comparing WT and CALR DEL HOM cluster 3 (HSC) (blue) and cluster 11 (MK) (red), as measured by combining the z scores from WT and MK analyses.
While cell cycle and UPR have previously been described as up-regulated in human CD34+ cells with CALR mutation (34), the discovery of cholesterol biosynthesis was somewhat unexpected. However, this aligned with the predicted significant activation of the lipid and cholesterol biosynthetic transcriptional machinery controlled by the sterol regulatory elementbinding proteins (SREBPs; SREBF1 and SREBF2) and the SREBF chaperone (SCAP) and their inhibitor insulin-induced gene 1 (INSIG1) (Fig. 6F). Moreover, as discussed further below, a role for cholesterol biosynthesis in a proliferative, platelet-biased blood disorder is biologically plausible. Upstream regulator analysis also pointed to activation of ERN1 (Ire1) and Xbp1, two constituents of UPR, as well as STAT5 (table S3), which is consistent with previous demonstrations that mutant CALR acts via STAT signaling (4, 3537). We additionally observed other previously undescribed signaling processes to be predicted to be activated, including drivers of proliferation such as CSF2 [granulocyte-macrophage colony-stimulating factor (GM-CSF)] and hepatocyte growth factor (HGF), or repressed, like the known tumor suppressors TP53 and let-7.
Single-cell transcriptomic approaches have allowed detailed examinations of differentiation landscapes in both normal and perturbed hematopoiesis without a requirement to initially define populations based on a set of cell surface markers. We therefore used single-cell transcriptomics to investigate our recently generated mutant CALR-driven mouse model of ET and found an expected increase in both HSCs and MK lineage cells. We also found an increase in a previously unknown group of cells, here termed pMKPs, linking HSCs with the MK lineage. In vitro, pMKPs displayed behaviors intermediate to those of HSCs and MkPs: Similarly to HSCs, they had some proliferative potential, but similarly to MkPs, they were almost exclusively restricted to the MK lineage. In transplantations, pMKPs and MkPs showed similar behavior: They both transiently produced platelets at a low level. We hypothesize that while pMKPs are more proliferative than MkPs in vitro, neither population is capable of sufficient proliferation to significantly contribute to platelet production in the transplant setting. While this manuscript was in preparation, another group described separating SLAM (Lin CD48 CD150+) cells based on EPCR and CD34, finding that EPCR SLAM cells performed poorly in transplants and showed gene expression profiles (high Gata1, Vwf, and Itga2b) indicative of MK bias (38), results that are broadly consistent with our own.
Our characterization of pMKPs accords well with an increasing understanding that at least a portion of megakaryopoiesis occurs via an early branch point directly from HSCs. While the standard model of hematopoiesis shows megakaryocytes subsequent to MPP2, lineage tracing experiments have shown that some MkPs are generated in an MPP2-independent way (19). Furthermore, in vivo labeling of the most primitive HSCs showed that within 1 week of label induction in LT-HSCs, label can be seen in MK lineages but no other, indicating that the HSC-to-MK pathway can be noticeably faster than pathways producing other lineages (22). Our results suggest that pMKPs are likely to arise independently of the MPP2 stage, as suicidal depletion of the earliest HSPCs reduces pMKPs to a much greater extent than MPP2s. It is therefore tempting to speculate that our pMKP sort scheme may isolate intermediate cells on this shorter, faster bypass trajectory. A recent study of JAK2 V617F-driven MF in humans attributed increased megakaryopoiesis to the expansion of both traditional MkPs and a novel MkP-like population, suggesting that cells that may be analogous to our pMKPs are relevant in human disease (30).
We also investigated an outstanding question about at which stages mutant CALR acts to drive a platelet phenotype. Mutant CALR has been demonstrated to increase the number of immunophenotypic HSCs and MkPs (6), and we also saw an expansion in the number of pMKPs. When considering the behavior of cells individually, it is clear that mutant CALR acts from the stem cell compartment: CALR DEL HOM HSCs were more proliferative and faster to produce megakaryocytes than were their WT counterparts. Mutant CALR did not show a strong effect on the proliferation or MK bias of pMKPs at the level of a single cell but drove an increase in proliferation of MkPs and thus the number of megakaryocytes produced. We therefore concluded that mutant CALR drives platelet bias and proliferation at multiple stages of megakaryopoiesis, although this effect is strongest within HSCs.
Last, we used our single-cell transcriptomic data to ask which biological pathways were most differentially regulated in our CALR DEL HOM mice. Mutant CALR was associated with an up-regulation of the unfolded protein response, as would be expected for cells with impaired chaperone activity and as has been seen in human patient cells (34). In addition, mutant CALR cells showed an increase in cell cycle genes, again consistent with observations from human patient cells (34) and in agreement with our in vitro data, which showed that mutant CALR HSCs and MkPs were more proliferative. We also found up-regulation of cholesterol biosynthesis pathway genes in mutant CALR hematopoietic cells. While cholesterol biosynthesis is broadly increased across numerous cancers (39), including hematological cancers (40), CALR has also been directly linked to cholesterol biosynthesis. CALR/ mouse embryonic fibroblasts show impaired endoplasmic reticulum (ER) Ca2+ levels, leading to overactivation of SREBPs, which then up-regulate cholesterol and triacylglycerol biosynthesis genes (41). As mutant CALR lacks its Ca2+-binding domain, it is possible that CALR DEL HOM cells phenocopy knockout cells with respect to ER Ca2+ stores, thus leading to the observed overactive transcription of cholesterol biosynthesis genes. While megakaryocytes derived from human patient samples have been shown to have increased store-operated Ca2+ entry due to the perturbation of a complex between STIM1, ERp57, and CALR (42), none of our differentially activated pathways from IPA pointed to altered cytoplasmic Ca2+ signaling in the stem and progenitor populations tested. This may reflect differences between progenitor and mature cells. Mice with impaired cholesterol efflux have more proliferative HSCs (43) and an increase in MkP proliferation and an ET-like phenotype (44), suggesting that there may be a previously unknown link between the CALR DEL mutation, cholesterol metabolism, proliferation of MkPs, and thus the overproduction of platelets. While cholesterol biosynthesis was the most prominent novel target found in our transcriptomic analysis, it was by no means alone. IPA upstream regulator analysis predicted an up-regulation of interleukin-5 (IL-5), GM-CSF, and HGFall with known roles in hematopoiesisin addition to several unexpected results, such as TBX2, a transcription factor that has not been studied in hematopoiesis. Upstream regulators predicted to be decreased include the tumor suppressor TP53; let-7, a microRNA with a role in the self-renewal of fetal HSCs (45); and KDM5B (Jarid1b), a histone methylase required for HSC self-renewal (46).
Overall, our study has characterized a previously undescribed MK trajectory implicated in the progression of ET. We find that pMKPs are an intermediate stage within one pathway of megakaryopoiesis and hypothesize that they may be situated within the MPP2-independent MK shortcut. Last, our analysis confirmed that JAK-STAT signaling, unfolded protein response, and cell cycle are all increased by the presence of mutant CALR and found up-regulation of cholesterol biosynthesis, in addition to numerous other potential upstream regulators. Functional validation of these biological pathways and upstream regulators may represent promising avenues of future research to better understand mutant CALR-driven disease and in the development of therapeutic strategies.
The objectives of the study were to generate transcriptomic data from our CALR mouse model of ET and to use these data to determine how the hematopoietic landscape is affected by the CALR DEL mutation. All mouse procedures were performed in strict accordance with the U.K. Home Office regulations for animal research under project license 70/8406.
Bone marrow cells were harvested from the femurs, tibia, and iliac crests of mice. Bones were crushed in a mortar and pestle in phosphate-buffered saline (PBS) and 2% fetal bovine serum (FBS) and 5 mM EDTA and then filtered through a 70-m filter to obtain a suspension of bone marrow cells. The suspension was incubated with an equal volume of ammonium chloride solution (STEMCELL Technologies, Vancouver, Canada) for 10 min on ice to lyse erythrocytes, followed by centrifugation for 5 min at 350g. The cell pellet was resuspended in PBS and 2% FBS and 5 mM EDTA, filtered again through a 70-m filter, and centrifuged again for 5 min at 350g. For cell sorting experiments, bone marrow mononuclear cell suspensions were immunomagnetically depleted of lineage (Lin)positive cells (EasySep Mouse Hematopoietic Progenitor Cell Isolation Kit, catalog no. 19856, STEMCELL Technologies). For staining, cells were incubated with the indicated antibodies for 40 min on ice; see attached tables for catalog information and concentrations used (table S4). Flow cytometry was performed on BD LSRFortessa analyzers, and flow cytometric sorting was performed on BD Influx 4 and 5 cell sorters (BD Biosciences, San Jose, USA). Flow data were analyzed using FlowJo software (Tree Star, Ashland, USA).
For 10x Chromium (10x Genomics, Pleasanton, CA) experiments, Lin c-Kit+ (LK) and Lin Sca1+ cKit+ (LSK) cells were sort purified as described above and processed according to the manufacturers protocol. Sample demultiplexing, barcodes processing, and gene counting were performed using the count commands from the Cell Ranger v1.3 pipeline (https://support.10xgenomics.com/single-cell-gene-expression/software/overview/welcome). After Cell Ranger processing, each sample (LK and LSK for WT and CALR HOM DEL) was filtered for potential doublets by simulating synthetic doublets from pairs of scRNAseq profiles and assigning scores based on a k nearest-neighbor classifier on PCA-transformed data. The 1 and 4.5% of cells with the highest doublets scores from each LSK or LK sample were removed from further analysis, respectively. Cells with >10% of unique molecular identifier (UMI) counts mapping to mitochondrial genes, expressing fewer than 500 genes, or with a total number of UMI counts further than 3 SDs from the mean were excluded. After quality control, 11,098 WT (5139 LK and 5959 LSK) and 15,547 HOM (7815 LK and 7732 LSK) cells were retained for downstream analysis from our first repeat. For our second repeat, 3451 WT (2479 LK and 972 LSK) and 12,372 HOM (7824 LK and 4548 LSK) cells were retained for downstream analysis. These cells were then normalized to the same total count. All scRNAseq data were analyzed using the Scanpy Python Module (47).
To assign cell type identities to WT and CALR samples, a previously published landscape of 45,000 WT LK and LSK hematopoietic progenitors (24) was used as a reference for cell type annotation. This reference was clustered using Louvain clustering, resulting in 13 clusters. LK + LSK samples were joined for each genotype (WT and CALR DEL HOM) and projected into the PCA space of this reference dataset. Nearest neighbors were calculated between the two datasets based on Euclidean distance in the top 50 PCA components. Cells were assigned to the same cluster to which the majority of their 15 nearest neighbors in the reference belonged.
A force-directed graph visualization of the 45,000 cell reference dataset was calculated by first constructing a k = 7 nearest-neighbor graph from the data, which was then used as input for the ForceAtlas2 algorithm as implemented in Gephi 0.9.1 (https://gephi.org). In the ForceAtlas2 algorithm, all cells are pushed away from each other, with the nearest-neighbor connections pulling them back together to segregate separate trajectories.
A fine-resolution clustering of the reference dataset was calculated using the Louvain algorithm, resulting in 63 clusters. These were used as input for a PAGA analysis of the reference dataset using the Scanpy Python Module with default parameters. The results of the PAGA analysis were visualized by using the nodes and their edge weights as input into the ForceAtlas2 algorithm for calculating force-directed graphs as implemented in Gephi 0.9.1. For visualization, only connections with edge weights of >0.3 were shown.
To visualize gene expression of the PAGA graph, the mean normalized expression of all cells belonging to each node was calculated and displayed on a per-node basis.
To calculate differential abundances, votes were given out from each WT LK and CALR LK cell to their k-nearest neighbors in the reference dataset, with k chosen such that the total number of votes given out by each sample was the same. For each cell in the reference dataset, the difference between the number of votes received from the WT and CALR HOM samples was calculated. This difference acts as a proxy for the differential abundance of WT and CALR HOM cells for the region of the LK landscape in which the reference cell is located. This differential abundance proxy could then be visualized either on the reference landscape itself or on the PAGA graph calculated using the reference landscape. In the latter case, each node of the PAGA graph was colored by the mean differential abundance of all cells belonging to that node.
After flow sorting, cells were cultured in StemSpan SFEM (serum-free expansion medium) (STEMCELL Technologies) supplemented with 10% FBS (STEMCELL Technologies), 1% penicillin/streptomycin (Sigma-Aldrich), 1% l-glutamine (Sigma-Aldrich), stem cell factor (SCF; 250 ng/ml), IL-3 (10 ng/ml), and IL-6 (10 ng/ml; STEMCELL Technologies), with or without thrombopoietin (100 ng/ml; STEMCELL Technologies), in round-bottom 96-well plates (Corning, Corning, USA). For pro-erythroid conditions, cells were cultured as above but with the following cytokines: SCF (250 ng/ml), THPO (thrombopoietin) (50 ng/ml), EPO (erythropoietin) (5 U/ml), IL-3 (20 ng/ml), and Flt3L (50 ng/ml). For pro-myeloid conditions, cells were cultured as above but with the following cytokines: SCF (250 ng/ml), THPO (50 ng/ml), granulocyte colony-stimulating factor (50 ng/ml), IL-3 (20 ng/ml), Flt3L (50 ng/ml), and GM-CSF (50 ng/ml).
At 1, 2, 3, 4, and, in some cases, 7 days after flow sorting, single cellderived clones were visually inspected. Wells with surviving cells were classified into one of four categories: (i) exactly one enlarged cell, presumed to be a megakaryocyte; (ii) multiple enlarged cells; (iii) mixed expansion, with both small and enlarged cells; and (iv) expansion with only small cells. In some cases, the experimenter was blinded to the identity of the cell population initially sorted into the well he/she was inspecting and the genotype of the mouse.
For immunofluorescence, cells were allowed to adhere to the surface of poly-l-lysinecoated slides for 30 min at 37C (Poly-Prep Slides, Sigma-Aldrich). Cells were then fixed with 4% paraformaldehyde (Sigma-Aldrich) in PBS overnight at 4C, permeabilized with 0.25% Triton X-100 (Sigma-Aldrich) in PBS for 10 min at room temperature, and blocked with 1% bovine serum albumin (Sigma-Aldrich) for 1 hour at room temperature. Cells were stained with CD41 Alexa Fluor 488 (BioLegend, catalog no. 133908) overnight and mounted with 4,6-diamidino-2-phenylindole (DAPI) (VECTASHIELD Mounting Medium with DAPI, Vector Laboratories Inc., Burlingame, USA; catalog no. H-1500). Pictures were acquired on LSM-710 and LSM-780 confocal microscopes (Zeiss) and analyzed using ZEN software (Zeiss). For quantification of immunofluorescence, cells were cultured on CD44-coated glass-bottom plates for immobilization (48), followed by fixation and staining as above. Pictures were acquired on a Leica DMI4000 microscope (Leica), and CD41 intensity and cell size were quantified using Fiji software.
FACS-sorted cells from VWF-GFP+ donors were injected into the tail veins of W41/W41 (CD45.1) recipient that had been sublethally irradiated with 1 400 centigrays with 250,000 spleen cells as helpers. Peripheral blood was analyzed 1, 2, 3, 4, and 16 weeks after transplant for all cohorts.
Differential expression analysis was performed between WT (LK + LSK) and CALR DEL HOM (LK + LSK) clusters using the Wilcoxon rank sum test on all genes that passed initial quality control (typically approximately 15,000). A Benjamini-Hochberg correction was applied to correct for multiple testing. Genes with an adjusted P value of <0.05 and a fold change of >1.5 between genotypes were marked as differentially expressed. The original normalized counts were used in all cases.
DEGs were studied using IPA (Qiagen). We imputed the whole transcriptome in IPA and then filtered for analysis only statistically significant (adjusted P < 0.01) items with a log2FC > 0.3785 or log2FC < 0.3785. Pathways and upstream regulator networks showing relationships and interactions experimentally confirmed between DEGs and others that functionally interact with them were generated and ranked in terms of significance of participating genes (P < 0.05) and activation status (z score).
Data were analyzed, and graphs were generated in Microsoft Excel (Microsoft) and GraphPad PRISM (GraphPad, La Jolla, USA). Data are presented as means SD. Unless otherwise stated, statistical tests were unpaired Students t tests. P values are as follows: *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001.
Acknowledgments: We would like to acknowledge J. Aungier, T. Hamilton, D. Pask, and R. Sneade for invaluable technical assistance; R. Schulte, C. Cossetti, and G. Grondys-Kotarba at the CIMR Flow Cytometry Core Facility for assistance with cell sorting; and S. Loughran, T. Klampfl, and E. Laurenti for valuable discussions. Funding: Work in the Gttgens laboratory is supported by the Medical Research Council (MR/M008975/1), Wellcome (206328/Z/17/Z), Blood Cancer UK (18002), and Cancer Research UK (RG83389, jointly with A.R.G.). Work in the Green laboratory is supported by Wellcome (RG74909), WBH Foundation (RG91681), and Cancer Research UK (RG83389, jointly with B.G.). Author contributions: D.P. and H.J.P. designed and conducted experiments with assistance from J.L. S.W. and H.P.B. performed bioinformatic analyses. M.V. performed IPA with supervision from A.V.-P. A.G. provided DTA mice. D.P. analyzed data and wrote the manuscript with input from H.J.P. and J.L. and supervision from B.G. and A.R.G. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. We have deposited scRNAseq data in the NCBI Gene Expression Omnibus (GEO) database with accession number GSE160466. Additional data related to this paper may be requested from the authors.
Read the original here:
The stem/progenitor landscape is reshaped in a mouse model of essential thrombocythemia and causes excess megakaryocyte production - Science Advances
Recommendation and review posted by Bethany Smith
New Insights on the Role of the Mesenchymal-Hematopoietic Stem Cell Axis in Autologous and Allogeneic Hematopoiesis – DocWire News
This article was originally published here
Stem Cells Dev. 2020 Nov 24. doi: 10.1089/scd.2020.0148. Online ahead of print.
ABSTRACT
Cytoreductive protocols are integral both as conditioning regimens for bone marrow transplantation and as part of therapies for malignancies, but their associated comorbidities represent a long-standing clinical problem. In particular, they cause myeloablation that debilitates the physiological role of mesenchymal stem and precursor cells (MSPCs) in sustaining hematopoiesis. This review addresses the damaging impact of cytoreductive regimens on MSPCs. Additionally, it discusses prospects for alleviating the resulting iatrogenic comorbidities. New insights into the structural and functional dynamics of hematopoietic stem cell (HSC) niches reveal the existence of empty niches and the ability of the donor-derived healthy HSCs to outcompete the defective HSCs in occupying these niches. These findings support the notion that conditioning regimens, conventionally used to ablate the recipient hematopoiesis to create space for engraftment of the donor-derived HSCs, may not be a necessity for allogeneic bone marrow transplantation. Additionally, the capacity of the MSPCs to cross-talk with hematopoietic stem cells, despite MHC disparity, and suppress graft versus host disease indicates the possibility for development of a conditioning-free, MSPCs-enhanced protocol for bone marrow transplantation. The clinical advantage of supplementing cytoreductive protocols with MSPCs to improve autologous hematopoiesis reconstitution and alleviate cytopenia associated with chemo and radiation therapies for cancer is also discussed.
PMID:33231142 | DOI:10.1089/scd.2020.0148
Go here to read the rest:
New Insights on the Role of the Mesenchymal-Hematopoietic Stem Cell Axis in Autologous and Allogeneic Hematopoiesis - DocWire News
Recommendation and review posted by Bethany Smith